US10280398B2 - Midbrain dopamine (DA) neurons for engraftment - Google Patents

Midbrain dopamine (DA) neurons for engraftment Download PDF

Info

Publication number
US10280398B2
US10280398B2 US14/356,042 US201214356042A US10280398B2 US 10280398 B2 US10280398 B2 US 10280398B2 US 201214356042 A US201214356042 A US 201214356042A US 10280398 B2 US10280398 B2 US 10280398B2
Authority
US
United States
Prior art keywords
cells
cell
neurons
signaling
midbrain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US14/356,042
Other languages
English (en)
Other versions
US20150010514A1 (en
Inventor
Lorenz Studer
Jae-Won Shim
Sonja Kriks
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Memorial Sloan Kettering Cancer Center
Original Assignee
Memorial Sloan Kettering Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48192829&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US10280398(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Memorial Sloan Kettering Cancer Center filed Critical Memorial Sloan Kettering Cancer Center
Priority to US14/356,042 priority Critical patent/US10280398B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: SLOAN-KETTERING INSTITUTE FOR CANCER RES
Assigned to MEMORIAL SLOAN-KETTERING CANCER CENTER reassignment MEMORIAL SLOAN-KETTERING CANCER CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KRIKS, Sonja, SHIM, Jae-Won, STUDER, LORENZ
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: SLOAN-KETTERING INST CAN RESEARCH
Publication of US20150010514A1 publication Critical patent/US20150010514A1/en
Application granted granted Critical
Publication of US10280398B2 publication Critical patent/US10280398B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/16Activin; Inhibin; Mullerian inhibiting substance
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/41Hedgehog proteins; Cyclopamine (inhibitor)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/72Transferases (EC 2.)
    • C12N2501/727Kinases (EC 2.7.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells

Definitions

  • the present invention relates to the field of stem cell biology, in particular the lineage specific differentiation of pluripotent or multipotent stem cells, which can include, but is not limited to, human embryonic stem cells (hESC) in addition to nonembryonic human induced pluripotent stem cells (hiPSC), somatic stem cells, stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • hESC human embryonic stem cells
  • hiPSC human induced pluripotent stem cells
  • somatic stem cells stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • DA dopamine
  • the midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons made using the methods of the present invention are further contemplated for various uses including, but not limited to, use in in vitro drug discovery assays, neurology research, and as a therapeutic to reverse disease of, or damage to, a lack of dopamine neurons in a patient. Further, compositions and methods are provided for differentiating midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons from human pluripotent stem cells for use in disease modeling, in particular Parkinson's disease. Additionally, authentic DA neurons are enriched for markers, such as CD142, and A9 type neuronal cells.
  • Pluripotent cells may be from embryonic and/or nonembryonic somatic stem cell origin.
  • lineage specific differentiated cell populations are contemplated to find use in cell replacement therapies for patients with diseases resulting in a lose of function of a defined cell population.
  • lineage specific differentiated cells are also valuable research tools for a variety of purposes including in vitro screening assays to identify, confirm, and test for specification of function or for testing delivery of therapeutic molecules to treat cell lineage specific disease.
  • Previously embryonic and somatic stem cells were used as therapeutics and model systems for neurodegenerative diseases.
  • Research and technological developments relating to directed differentiation of embryonic and somatic stem cells has taken place in the field of diseases of the central nervous system (CNS), such as for Huntington's, Alzheimer's, Parkinson's, and multiple sclerosis.
  • CNS central nervous system
  • the results of these studies showed little capability of these cells used in vivo to allow the patient to recover neuronal function and often resulted in the growth of unwanted tumors in the patients.
  • compositions and methods to obtain cell populations capable of being used both in research and as a therapeutic for treating diseases resulting in a loss of cells having a particular function Therefore there is a need for compositions and methods to obtain cell populations capable of being used both in research and as a therapeutic for treating diseases resulting in a loss of cells having a particular function.
  • the present invention relates to the field of stem cell biology, in particular the lineage specific differentiation of pluripotent or multipotent stem cells, which can include, but is not limited to, human embryonic stem cells (hESC) in addition to nonembryonic human induced pluripotent stem cells (hiPSC), somatic stem cells, stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • hESC human embryonic stem cells
  • hiPSC human induced pluripotent stem cells
  • somatic stem cells stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • DA dopamine
  • the midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons made using the methods of the present invention are further contemplated for various uses including, but not limited to, use in in vitro drug discovery assays, neurology research, and as a therapeutic to reverse disease of, or damage to, a lack of dopamine neurons in a patient. Further, compositions and methods are provided for differentiating midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons from human pluripotent stem cells for use in disease modeling, in particular Parkinson's disease. Additionally, authentic DA neurons are enriched for markers, such as CD142, and A9 type neuronal cells.
  • a kit comprising a first signaling inhibitor, a second signaling inhibitor and a third signaling inhibitor, wherein said first inhibitor is capable of lowering transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling, said second inhibitor is capable of lowering Small Mothers against Decapentaplegic (SMAD) signaling and said third inhibitor is capable of lowering glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ) for activation of wingless (Wnt) signaling.
  • said first inhibitor is LDN-193189.
  • said first inhibitor is selected from the group consisting of LDN-193189, derivatives thereof and mixtures thereof.
  • said second inhibitor is SB431542.
  • said second inhibitor is selected from the group consisting of SB431542, derivatives thereof and mixtures thereof.
  • said third inhibitor is CHIR99021. In other embodiments, said third inhibitor is selected from the group consisting of CHIR99021, derivatives thereof and mixtures thereof.
  • said the kit further comprises an activator of Sonic hedgehog (SHH) signaling and an activator of fibroblast growth factor (FGF) 8 receptor family signaling. In one embodiment, said the kit further comprises brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP and transforming growth factor type ⁇ 3.
  • BDNF brain-derived neurotrophic factor
  • AA ascorbic acid
  • AA glial cell line-derived neurotrophic factor
  • dibutyryl cAMP transforming growth factor type ⁇ 3.
  • said the kit further comprise antibodies selected from the group consisting of anti-tyrosine hydroxylase (TH), anti-forkhead box protein A2 (FOXA2), and anti-LIM homeobox transcription factor 1, alpha (LMX1A).
  • said the kit further comprises a cell selected from the group consisting of a stem cell, embryonic stem cell, induced pluripotent stem cell, and an engineered cell.
  • said the kit further comprises instructions for differentiating progenitor cells and midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons.
  • said the kit further comprises instructions for obtaining a cell from a patient with Parkinson's disease (PD).
  • PD Parkinson's disease
  • a composition comprising, a cell population in contact with a first signaling inhibitor and a second signaling inhibitor, wherein greater than 40% of said cell population is positive for forkhead box protein A2 (FOXA2), wherein said cell population was previously contacted by a first signaling inhibitor, a third signaling inhibitor, and an activator of Sonic hedgehog (SHH) signaling, wherein said first inhibitor is capable of lowering transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling, said second inhibitor is capable of lowering glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ) signaling for activation of wingless (Wnt) signaling and said third inhibitor is capable of lowering Small Mothers against Decapentaplegic (SMAD) signaling.
  • FXA2 forkhead box protein A2
  • SHH Sonic hedgehog
  • said first inhibitor is a small molecule selected from the group consisting of LDN-193189, derivatives thereof and mixtures thereof.
  • said second inhibitor is selected from the group consisting of CHIR99021 and derivatives thereof.
  • said third inhibitor is selected from the group consisting of SB431542, derivatives thereof and mixtures thereof.
  • said activator of Sonic hedgehog (SHH) signaling is selected from the group consisting of Sonic hedgehog (SHH) C25II and smoothened (SMO) receptor small molecule agonist, wherein said agonist is purmorphamine.
  • said cell population was further previously contacted with Fibroblast growth factor 8 (FGF8).
  • FGF8 Fibroblast growth factor 8
  • said majority of cells comprising said cell population are forkhead box protein A2 (FOXA2) + LIM+ homeobox transcription factor 1+, alpha (LMX1A), + NGN2+ and DDC+ floor plate midbrain progenitor cells.
  • said cell population is selected from the group consisting of a rodent cells, primate cells and human cells.
  • said cells are derived from Parkinson's disease (PD) patient cells.
  • said cell population is at least 50% positive for forkhead box protein A2 (FOXA2).
  • said cell population is at least 60% positive for forkhead box protein A2.
  • said cell population is at least 70% positive for forkhead box protein A2.
  • said cell population is at least 80% positive for forkhead box protein A2. In one embodiment, said cell population is at least 90% positive for forkhead box protein A2. In one embodiment, said cell population is at least 95% up to 100% positive for forkhead box protein A2.
  • a composition comprising, an in vitro cell population wherein the majority of cells comprising said cell population are tyrosine hydroxylase (TH) + forkhead box protein A2 (FOXA2) + LIM homeobox transcription factor 1+, alpha (LMX1A) + floor plate midbrain dopamine (DA) neurons.
  • said greater than 40% of said floor plate midbrain dopamine (DA) neurons are tyrosine hydroxylase positive (TH+).
  • said cell population is at least 50% tyrosine hydroxylase positive.
  • said cell population is at least 60% tyrosine hydroxylase positive.
  • said cell population is at least 70% tyrosine hydroxylase positive.
  • said cell population is at least 80% tyrosine hydroxylase positive. In one embodiment, said cell population is at least 90% tyrosine hydroxylase positive. In one embodiment, said cell population is at least 95% up to 100% tyrosine hydroxylase positive. In some embodiments, said cell population comprises a majority of midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons.
  • DA dopamine
  • said floor plate midbrain dopamine (DA) neurons are positive for markers selected from the group consisting of nuclear receptor NURR1 (NR4A2), Neuron-specific class III beta-tubulin (Tuj1), TTF3, paired-like homeodomain 3 (PITX3), achaete-scute complex (ASCL), early B-cell factor 1 (EBF-1), early B-cell factor 3 (EBF-3) and transthyretin (TTR).
  • said midbrain fate FOXA2/LMX1A+ dopamine (DA) neuron population is positive for a molecule selected from the group consisting of DA, 3,4-Dihydroxy-Phenylacetic Acid (DOPAC) and homovanillic acid (HVA).
  • said marker is selected from the group consisting of a protein and a nucleic acid.
  • said midbrain fate FOXA2/LMX1A+ dopamine (DA) neuron population is capable of engrafting in vivo in a patient selected from the group consisting of a Parkinson disease (PD) patient.
  • said midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons are capable of engrafting in vivo and providing dopamine (DA) neuronal function.
  • the inventions provide a composition, comprising, a cell population in contact with LDN-193189 and CHIR99021, wherein greater than 40% of said cell population is positive for forkhead box protein A2 (FOXA2), and wherein said cell population was previously contacted by LDN-193189, SB431542, an activator of Sonic hedgehog (SHH) signaling and CHIR99021.
  • said activator of SONIC hedgehog (SHH) signaling is selected from the group consisting of Sonic hedgehog (SHH) C25II and purmorphamine.
  • said greater than 10% of said cell population is double positive for forkhead box protein A2 (FOXA2) and LIM homeobox transcription factor 1, alpha (LMX1A).
  • said majority of said cell population is a population of floor plate midbrain progenitor cells.
  • said cell population was previously contacted with fibroblast growth factor 8 (FGF8).
  • FGF8 fibroblast growth factor 8
  • said cell population is selected from the group consisting of rodent cells, primate cells and human cells.
  • said human cells are cells from a patient with a neurological symptom of Parkinson's disease (PD).
  • said cell population is derived from an induced pluripotent stem cell (iPSC).
  • greater than 10% of said cell population is selected from the group consisting of double positive for forkhead box protein A2 (FOXA2)/LIM homeobox transcription factor 1, alpha (LMX1A) and double positive for forkhead box protein A2 (FOXA2)/orthodenticle homeobox 2 (OTX2).
  • the inventions provide a method for inducing directed differentiation of cells into a population of floor plate midbrain progenitor cells, comprising, a) providing: i) a cell population, wherein said cell population is selected from the group consisting of a nonembryonic stem cell, an embryonic stem cell, an induced nonembryonic pluripotent cell and an engineered pluripotent cell; and ii) a first signaling inhibitor, a second signaling inhibitor, an activator of Sonic hedgehog (SHH) signaling and a third signaling inhibitor, wherein said first inhibitor is capable of lowering transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling, said second inhibitor is capable of lowering Small Mothers against Decapentaplegic (SMAD) signaling and said third inhibitor is capable of lowering glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ) signaling for activation of wingless (Wnt) signaling; b) contacting said cell population with said first and said second
  • said contact with said first and said second inhibitor is under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells. In one embodiment, said contact with said first and said second inhibitor is within 1 hour of plating cells in vitro. In one embodiment, said contact with said first and said second inhibitor is within 48 hours of plating cells in vitro. In one embodiment, said contact with said first and said second inhibitor is within 62 hours of plating cells in vitro. In one embodiment, said contact of said cells with said activator of Sonic hedgehog (SHH) signaling is under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells.
  • SHH Sonic hedgehog
  • said contact of said cells with said activator of Sonic hedgehog (SHH) signaling is at least 24 hours up to 36 hours after contacting said cell population with said first and said second inhibitor. In one embodiment, said contact of said cells with said activator of Sonic hedgehog (SHH) signaling is up to 144 hours. In one embodiment, said contact of said cells with said third inhibitor is under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells. In one embodiment, said contact of said cells with said third inhibitor is at least 24 hours up to 36 hours after contacting said cell population with said activator of Sonic hedgehog (SHH) signaling. In one embodiment, said contact of said cells with said third inhibitor is up to 192 hours.
  • said cell population is differentiated into said floor plate midbrain progenitor cells by at least day 11 after contacting said cells with said first and said second inhibitor.
  • said first inhibitor is SB431542.
  • said second inhibitor is LDN-193189.
  • said third inhibitor is CHIR99021.
  • said activator of Sonic hedgehog (SHH) signaling is selected from the group consisting of Sonic hedgehog (SHH) C25II and purmorphamine.
  • said method further provides Fibroblast growth factor 8 (FGF8) and contacting said cell population with said FGF8 under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells.
  • FGF8 Fibroblast growth factor 8
  • said contact of said cells with said FGF8 is at least 24 up to 36 hours after contacting said cell population with said first and said second inhibitor. In one embodiment, said contact of said cells with said FGF8 is up to 144 hours. In one embodiment, said floor plate midbrain progenitor cell population comprises greater than 40% forkhead box protein A2 (FOXA2) + cells. In one embodiment, said floor plate midbrain progenitor cell population comprises greater than 40% forkhead box protein A2 (FOXA2) + LIM homeobox transcription factor 1, alpha (LMX1A) + cells.
  • said method further comprises step e) contacting said population of floor plate midbrain progenitor cells with neuronal maturation medium, said medium comprising N2 medium, brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP (dbcAMP) and transforming growth factor type ⁇ 3 for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • said method further comprises step e) contacting said population of floor plate midbrain progenitor cells with neuronal maturation medium with B27 supplement for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • said cells contacted with neurobasal medium with B27 supplement are contacted with brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP (dbcAMP) and transforming growth factor type ⁇ 3 for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • BDNF brain-derived neurotrophic factor
  • AA ascorbic acid
  • dbcAMP dibutyryl cAMP
  • transforming growth factor type ⁇ 3 for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • said floor plate midbrain dopamine (DA) neurons are forkhead box protein A2 (FOXA2) + LIM homeobox transcription factor 1, alpha (LMX1A) + , Nuclear receptor related 1 protein (NURR1) + and tyrosine hydroxylase (TH) + .
  • greater than 40% of said floor plate midbrain dopamine (DA) neurons are tyrosine hydroxylase (TH) + .
  • said population of floor plate midbrain dopamine (DA) neurons are differentiated by at least day 25 after contacting said cell population with said first and said second inhibitor.
  • said floor plate midbrain dopamine (DA) neurons are positive for markers that identify molecules.
  • said markers are selected from the group consisting of tyrosine hydroxylase (TH), forkhead box protein A2 (FOXA2), LIM homeobox transcription factor 1, dompamine, 3,4-Dihydroxy-Phenylacetic Acid (DOPAC) and homovanillic acid (HVA), alpha, nuclear receptor NURR1 (NR4A2), Neuron-specific class III beta-tubulin (Tuj1), TTF3, paired-like homeodomain 3 (PITX3), achaete-scute complex (ASCL), early B-cell factor 1 (EBF-1), early B-cell factor 3 (EBF-3), transthyretin (TTR), synapsin, dopamine transporter (DAT), and G-protein coupled, and inwardly rectifying potassium channel (Kir3.2/GIRK2).
  • TH tyrosine hydroxylase
  • FOXA2 forkhead box protein A2
  • DOPAC 3,4-Dihydroxy-Phenylacetic Acid
  • HVA homovanil
  • said molecule is selected from the group consisting of a protein and a nucleic acid. In one embodiment, said molecule is identified using a marker selected from the group consisting of an antibody, a PCR primer, a nucleic acid sequence and an enzyme assay.
  • said floor plate midbrain dopamine (DA) neurons are capable of engrafting in vivo in a patient with Parkinson disease (PD) for providing dopamine (DA) neuronal function.
  • said method further comprises, providing, a patient in need of dopamine producing neurons, wherein said patient shows at least one neurological symptom, and the step of transplanting floor plate midbrain dopamine (DA) neurons into said patient for providing dopamine (DA) neuronal function.
  • said neurological symptoms are selected from the group consisting of tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability, and rigidity.
  • said patient shows a reduction of said neurological symptom.
  • said cell is selected from a rodent cell, a primate cell and a human cell.
  • said human cells are cells from a patient with a neurological symptom of Parkinson's disease (PD).
  • PD Parkinson's disease
  • the inventions provide a method of engrafting in vivo for therapeutic treatment, comprising, a) providing: i) a population of floor plate midbrain dopamine (DA) neurons wherein greater than 40% of said population expresses tyrosine hydroxylase (TH); and ii) a subject, wherein said subject shows at least one neurological symptom; and b) transplanting said floor plate midbrain dopamine (DA) neurons into said subject under conditions for allowing in vivo engraftment for providing dopamine (DA) neuronal function.
  • said neurological symptoms are selected from the group consisting of tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability and rigidity.
  • said subject shows reduction of said neurological symptom.
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a population of floor plate midbrain progenitor cells treated according to methods of the present inventions.
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a population of floor plate midbrain progenitor cells treated according to a method further comprising a step of contacting said population of floor plate midbrain progenitor cells with neuronal maturation medium, said medium comprising N2 medium, brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP and transforming growth factor type ⁇ 3 for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • BDNF brain-derived neurotrophic factor
  • AA ascorbic acid
  • glial cell line-derived neurotrophic factor dibutyryl cAMP
  • said population of floor plate midbrain progenitor cells are derived from a cell population treated according to a method of the present inventions.
  • said population of floor plate midbrain progenitor cells are derived from a cell population treated according to a method for inducing directed differentiation of cells into a population of floor plate midbrain progenitor cells, comprising, a) providing: i) a cell population, wherein said cell population is selected from the group consisting of a nonembryonic stem cell, an embryonic stem cell, an induced nonembryonic pluripotent cell and an engineered pluripotent cell; and ii) a first signaling inhibitor, a second signaling inhibitor, an activator of Sonic hedgehog (SHH) signaling and a third signaling inhibitor, wherein said first inhibitor is capable of lowering transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling, said second inhibitor is capable of lowering Small Mothers against Decapentaplegic (SMAD) signaling and said
  • TGF ⁇
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a cell population selected from the group consisting of animals, primates and humans.
  • said human cells are cells from a patient with a symptom of Parkinson's disease (PD).
  • PD Parkinson's disease
  • the inventions provide a composition, comprising, a cell population in contact with LDN-193189 and CHIR99021, wherein greater than 40% of said cell population is positive for forkhead box protein A2 (FOXA2), and wherein said cell population was previously contacted by LDN-193189, SB431542, an activator of Sonic hedgehog (SHH) signaling and CHIR99021, wherein said activator of Sonic hedgehog (SHH) signaling is selected from the group consisting of Sonic hedgehog (SHH) C25II and purmorphamine.
  • said cell population is at least 50% positive for forkhead box protein A2 (FOXA2).
  • said cell population is at least 60% positive for forkhead box protein A2.
  • said cell population is at least 70% positive for forkhead box protein A2. In one embodiment, said cell population is at least 80% positive for forkhead box protein A2. In one embodiment, said cell population is at least 90% positive for forkhead box protein A2. In one embodiment, said cell population is at least 95% up to 100% positive for forkhead box protein A2. In one embodiment, greater than 10% of said cell population is selected from the group consisting of double positive for forkhead box protein A2 (FOXA2)/LIM homeobox transcription factor 1, alpha (LMX1A) and double positive for forkhead box protein A2 (FOXA2)/orthodenticle homeobox 2 (OTX2).
  • FOXA2 double positive for forkhead box protein A2
  • LMX1A alpha
  • OTX2 double positive for forkhead box protein A2
  • said cell population is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, at least 95% up to 100% positive. In one embodiment, at least 20% of said cell population is positive for a marker selected from the group consisting of Nurr1+, CD142, DCSM1, CD63 and CD99. In some embodiments, said cell population is at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, at least 95% up to 100% positive. In some embodiments, said cell population is at least 50%, 60%, 70%, 80%, 90%, at least 95% up to 100% positive. In one embodiment, said cell population is selected from the group consisting of rodent cells, primate cells, human cells and human cells from a patient with a neurological symptom of Parkinson's disease (PD).
  • PD Parkinson's disease
  • the inventions provide a method for inducing directed differentiation of cells into a population of floor plate midbrain progenitor cells, comprising, a) providing: i) a cell population, wherein said cell population is selected from the group consisting of a nonembryonic stem cell, an embryonic stem cell, an induced nonembryonic pluripotent cell and an engineered pluripotent cell; and ii) a first signaling inhibitor, a second signaling inhibitor, an activator of Sonic hedgehog (SHH) signaling and a third signaling inhibitor, wherein said first inhibitor is SB431542, said second inhibitor is LDN-193189, said activator of Sonic hedgehog (SHH) signaling is selected from the group consisting of Sonic hedgehog (SHH) C25II and a purmorphamine, and said third inhibitor is CHIR99021; b) contacting said cell population with said first and said second inhibitor, wherein said contact with said first and said second inhibitor is under conditions capable of resulting in said differentiated population of floor plate midbrain
  • contact of said cells with said activator of Sonic hedgehog (SHH) signaling is under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells such that said contact of said cells with said activator of Sonic hedgehog (SHH) signaling is at least 24 hours and up to 36 hours after contacting said cell population with said first and said second inhibitor.
  • contact of said cells with said third inhibitor is under conditions capable of resulting in said differentiated population of floor plate midbrain progenitor cells such that said contact of said cells with said third inhibitor is at least 24 hours and up to 36 hours after contacting said cell population with said activator of Sonic hedgehog (SHH) signaling.
  • floor plate midbrain progenitor cell population comprises greater than 40% forkhead box protein A2 (FOXA2) + LIM homeobox transcription factor 1+, alpha (LMX1A) + cells.
  • the method further comprises step e) contacting said population of floor plate midbrain progenitor cells with neuronal maturation medium, said medium comprising N2 medium, brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP and transforming growth factor type ⁇ 3 for differentiation of floor plate midbrain progenitor cells into floor plate midbrain dopamine (DA) neurons.
  • BDNF brain-derived neurotrophic factor
  • AA ascorbic acid
  • glial cell line-derived neurotrophic factor dibutyryl cAMP
  • DA floor plate midbrain dopamine
  • said floor plate midbrain dopamine (DA) neurons are positive for sets of markers selected from the group consisting of forkhead box protein A2 (FOXA2) / LIM homeobox transcription factor 1 alpha (LMX1A)/tyrosine hydroxylase (TH); forkhead box protein A2/LIM homeobox transcription factor 1 alpha/tyrosine hydroxylase/CD142; forkhead box protein A2/LIM homeobox transcription factor 1 alpha/tyrosine hydroxylase/Nuclear receptor related 1 protein (NURR1); forkhead box protein A2/LIM homeobox transcription factor 1 alpha/tyrosine hydroxylase/CD142/Nuclear receptor related 1 protein and tyrosine hydroxylase/ ⁇ -synuclein.
  • FXA2 forkhead box protein A2
  • LIM homeobox transcription factor 1 alpha LIM homeobox transcription factor 1 alpha
  • TH LIM homeobox transcription factor 1 alpha/tyrosine hydroxylase
  • NURR1 Nuclear
  • said method further comprises step f) sorting said floor plate midbrain dopamine (DA) neurons for CD142 expression into a population of cells at least 80% positive for CD142.
  • said floor plate midbrain dopamine (DA) neurons are positive for a marker that identifies a molecule selected from the group consisting of tyrosine hydroxylase (TH), forkhead box protein A2 (FOXA2), LIM homeobox transcription factor 1, dompamine, 3,4-Dihydroxy-Phenylacetic Acid (DOPAC) and homovanillic acid (HVA), alpha, nuclear receptor NURR1 (NR4A2), Neuron-specific class III beta-tubulin (Tuj1), TTF3, paired-like homeodomain 3 (PITX3), achaete-scute complex (ASCL), early B-cell factor 1 (EBF-1), early B-cell factor 3 (EBF-3), transthyretin (TTR), synapsin, dopamine transporter (DAT),
  • TH
  • the method further comprises, provides, a patient in need of dopamine producing neurons, and a step after e) treating said patient by transplanting said floor plate midbrain dopamine (DA) neurons for providing dopamine (DA) neuronal function.
  • said patient comprises at least one neurological symptom selected from the group consisting of tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability, and rigidity.
  • said patient is observed to have at least one neurological symptom selected from the group consisting of tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability, and rigidity.
  • said patient shows a reduction of at least one of said neurological symptom.
  • the inventions provide a method of engrafting in vivo for therapeutic treatment, comprising, a) providing: i) a population of floor plate midbrain dopamine (DA) neurons wherein greater than 40% of said population expresses tyrosine hydroxylase (TH); and ii) a subject, wherein said subject shows at least one neurological symptom, wherein said neurological symptoms are selected from the group consisting of tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability and rigidity; and b) transplanting said floor plate midbrain dopamine (DA) neurons into said subject under conditions for allowing in vivo engraftment for providing dopamine (DA) neuronal function.
  • DA floor plate midbrain dopamine
  • said subject shows reduction of at least one of said neurological symptom.
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a population of cells further comprising a step of sorting said floor plate midbrain dopamine (DA) neurons for CD142 expression into a population of cells at least 80% positive for CD142.
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a population of floor plate midbrain progenitor cells after a step of sorting said floor plate midbrain dopamine (DA) neurons for CD142 expression into a population of cells at least 80% positive for CD142.
  • said population of floor plate midbrain dopamine (DA) neurons are derived from a cell population selected from the group consisting of animals, primates, humans and a patient with a symptom of Parkinson's disease (PD). In one embodiment, said population of floor plate midbrain dopamine (DA) neurons are derived from a cell population isolated from the group consisting of animals, primates, humans and a patient with a symptom of Parkinson's disease (PD).
  • PD Parkinson's disease
  • the term “disease modeling” refers to the process of using an experimental organism or in vitro cell cultures to mimic specific signs or symptoms observed in humans as a result of a disorder.
  • pluripotent stem cells derived from an animal model with a genetic mutation resulting in a neurological disorder such as Parkinson's disease (PD)
  • PD Parkinson's disease
  • human pluripotent stem cells derived from a person with a genetic mutation resulting in a neurological disorder such as Parkinson's disease (PD) can be grown and differentiated into neural cells harboring a similar defect observed within the person.
  • parkinsonism refers to a group of diseases that are all linked to an insufficiency of dopamine in the basal ganglia which is a part of the brain that controls movement. Symptoms include tremor, bradykinesia (extreme slowness of movement), flexed posture, postural instability, and rigidity. A diagnosis of parkinsonism requires the presence of at least two of these symptoms, one of which must be tremor or bradykinesia. The most common form of parkinsonism is idiopathic, or classic, Parkinson's disease (PD), but for a significant minority of diagnoses, about 15 percent of the total, one of the Parkinson's plus syndromes (PPS) may be present.
  • PD Parkinson's disease
  • PPS Parkinson's plus syndromes
  • Parkinson's disease involves the malfunction and death of vital nerve cells in the brain primarily in an area of the brain called the substantia nigra. Many of these vital nerve cells make dopamine, that as these neurons die off, the amount of dopamine resulting from differentiation in the brain decreases, leaving a person unable to control movement normally.
  • the intestines also have dopamine cells that degenerate in Parkinson's disease patients, and this may be an important causative factor in the gastrointestinal symptoms that are part of the disease. A group of symptoms that an individual experiences varies from person to person.
  • Primary motor signs of Parkinson's disease include the following: tremor of the hands, arms, legs, jaw and face, bradykinesia or slowness of movement, rigidity or stiffness of the limbs and trunk and postural instability or impaired balance and coordination.
  • the term “subject” refers to a mammal (human and animal, i.e. non-human animals) that is to be the recipient of a particular treatment including any type of control.
  • the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
  • non-human animals refers to all non-human animals including, but are not limited to, vertebrates such as rodents, non-human primates, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, canines, felines, ayes, etc.
  • dopamine refers to a chemical made by dopamine neurons that sends messages to the part of the brain containing neurons that control movement and coordination.
  • LSB refers to a combination of two compounds LDN-193189 and SB431542 capable of lowering or blocking signaling consisting of transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling and Small Mothers against Decapentaplegic (SMAD) signaling in a cell.
  • TGF ⁇ transforming growth factor beta
  • SAD Small Mothers against Decapentaplegic
  • SB431542 refers to a molecule capable of lowering or blocking transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling with a number CAS 301836-41-9, a molecular formula of C 22 H 18 N 4 O 3 , and a name of 4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]-benzamide, for example, see structure below:
  • SB431542 is StemoleculeTM SB431542, Stemgent, Inc. Cambridge, Mass., United States.
  • LDN-193189 refers to a small molecule DM-3189, IUPAC name 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline, with a chemical formula of C 25 H 22 N 6 .
  • LDN-193189 is capable of functioning as a SMAD signaling inhibitor.
  • LDN-193189 is also a highly potent small-molecule inhibitor of ALK2, ALK3, and ALK6, protein tyrosine kinases (PTK), inhibiting signaling of members of the ALK1 and ALK3 families of type I TGF ⁇ receptors, resulting in the inhibition of the transmission of multiple biological signals, including the bone morphogenetic proteins (BMP) BMP2, BMP4, BMP6, BMP7, and Activin cytokine signals and subsequently SMAD phosphorylation of Smad1, Smad5, and Smad8 (Yu et al. (2008) Nat Med 14:1363-1369; Cuny et al. (2008) Bioorg. Med. Chem. Lett. 18: 4388-4392, herein incorporated by reference).
  • BMP bone morphogenetic proteins
  • LDN-193189 is StemoleculeTM LDN-193189, Stemgent, Inc. Cambridge, Mass., United States.
  • GSK3 ⁇ inhibitor refers to a compound that inhibits a glycogen synthase kinase 3 ⁇ enzyme, for example, see, Doble, et al., J Cell Sci. 2003; 116:1175-1186, herein incorporated by reference.
  • a GSK3 ⁇ inhibitor is capable of activating a WNT signaling pathway, see, for example, Cadigan, et al., J Cell Sci. 2006; 119:395-402; Kikuchi, et al., Cell Signaling. 2007; 19:659-671, herein incorporated by reference.
  • CHIR99021 or “CHIR” or “aminopyrimidine” or “3-[3-(2-Carboxyethyl)-4-methylpyrrol-2-methylidenyl]-2-indolinone” refers to IUPAC name 6-(2-(4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl)pyrimidin-2-ylamino)ethylamino)nicotinonitrile.
  • GSK3 ⁇ glycogen synthase kinase 3 ⁇
  • CHIR99021 is StemoleculeTM CHIR99021, Stemgent, Inc. Cambridge, Mass., United States.
  • purmorphamine refers to a purine derivative, such as CAS Number: 483367-10-8, for one example see structure below, that activates the Hedgehog pathway including by targeting Smoothened.
  • purmorphamine is StemoleculeTM Purmorphamine, Stemgent, Inc. Cambridge, Mass., United States.
  • signal transduction protein refers to proteins that are activated or otherwise affected by ligand binding to a membrane receptor protein or some other stimulus.
  • Examples of signal transduction protein include a SMAD, a WNT complex protein, in another embodiment a WNT complex protein including beta-catenin, Sonic hedgehog (SHH), NOTCH, transforming growth factor beta (TGF ⁇ ), Activin, Nodal, glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ) proteins and the like.
  • SMAD SMAD
  • WNT complex protein in another embodiment a WNT complex protein including beta-catenin, Sonic hedgehog (SHH), NOTCH, transforming growth factor beta (TGF ⁇ ), Activin, Nodal, glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ) proteins and the like.
  • TGF ⁇ transforming growth factor beta
  • GSK3 ⁇ glycogen synthase kinase 3 ⁇
  • the ligand activated receptor must first interact with other proteins inside the cell before the ultimate physiological effect of the ligand on the cell's behavior is produced. Often, the behavior of a chain of several interacting cell proteins is altered following receptor activation or inhibition. The entire set of cell changes induced by receptor activation is called a signal transduction mechanism or signaling pathway.
  • LDN/S/F8/CHIR refers to contacting cells with LDN-193189 and SB431542 (i.e. LSB) in addition to S, Sonic Hedgehog activator, F8, FGF8, and CHIR of the present inventions.
  • LDN/SB refers to contacting cells with LDN-193189 and SB431542 (i.e. LSB) in addition to S, Sonic Hedgehog activator, F8, FGF8 but without CHIR as in previously published methods.
  • LDN/SB refers to contacting cells with LDN, LDN-193189 and SB, SB431542.
  • the term “inhibit” or “block” means a reduction in the level of activity of a particular signaling pathway of a cell upon treatment with a compound (i.e. an inhibitor) compared to the activity of said signaling pathway of a cell that is left untreated with such compound or treated with a control.
  • activate means an increase in the level of activity of a particular signaling pathway of a cell upon treatment with a compound (i.e. an activator) compared to the activity of said signaling pathway of a cell that is left untreated with such compound or treated with a control. Any level of inhibition or activation of a particular signaling pathway is considered an embodiment of the invention if such inhibition or activation results in the directed differentiation of a stem cell.
  • Sma Mothers Against Decapentaplegic or “Small Mothers against Decapentaplegic” or “SMAD” refers to a signaling molecule.
  • WNT or “wingless” in reference to a ligand refers to a group of secreted proteins (i.e. Int1 (integration 1) in humans) capable of interacting with a WNT receptor, such as a receptor in the Frizzled and LRPDerailed/RYK receptor family.
  • Int1 integration 1
  • a WNT receptor such as a receptor in the Frizzled and LRPDerailed/RYK receptor family.
  • WNT or “wingless” in reference to a signaling pathway refers to a signal pathway composed of Wnt family ligands and Wnt family receptors, such as Frizzled and LRPDerailed/RYK receptors, mediated with or without ⁇ -catenin.
  • a preferred WNT signaling pathway includes mediation by ⁇ -catenin, i.e. WNT/ ⁇ -catenin.
  • canonical pathway or “classical activation” in reference to WNT refers to one of the multiple Wnt downstream signal pathways, for example, in the canonical pathway a major effect of Wnt ligand binding to its receptor is the stabilization of cytoplasmic beta-catenin through inhibition of the beta-catenin degradation complex. Others Wnt pathways are non-canonical.
  • the small molecule CHIR affects a canonical Wnt signaling downstream pathway.
  • SHH Sonic hedgehog
  • Shh refers to a protein that is one of at least three proteins in the mammalian signaling pathway family called hedgehog, another is desert hedgehog (DHH) while a third is Indian hedgehog (IHH).
  • Shh interacts with at least two transmembrane proteins by interacting with transmembrane molecules Patched (PTC) and Smoothened (SMO).
  • PTC transmembrane molecules Patched
  • SMO Smoothened
  • Shh typically binds to PCT which then allows the activation of SMO as a signal transducer.
  • PTC transmembrane molecules Patched
  • SMO Smoothened
  • activator refers to small molecules, peptides, proteins and compounds for activating molecules resulting in directed differentiation of cells of the present inventions.
  • exemplary activators include but are not limited to: CHIR, Sonic hedgehog (SHH) C25II, a small molecule Smoothened agonist purmorphamine, fibroblast growth factor (FGF), etc.
  • the term “activator of Sonic hedgehog (SHH) signaling” refers to any molecule or compound that activates a SHH signaling pathway including a molecule or compound that binds to PCT or a Smoothened agonist and the like. Examples of such compounds are a protein Sonic hedgehog (SHH) C25II and a small molecule Smoothened agonist purmorphamine.
  • SHH C25II refers to a recombinant N-Terminal fragment of a full-length murine sonic hedgehog protein capable of binding to the SHH receptor for activating SHH, one example is R and D Systems catalog number: 464-5H-025/CF.
  • signals refer to internal and external factors that control changes in cell structure and function. They are chemical or physical in nature.
  • ligand refers to molecules and proteins that bind to receptors (R), examples include but are not limited to transforming growth factor-beta, activins, nodal, bone morphogenic proteins (BMPs), etc.
  • inhibitor or “signaling inhibitor” is in reference to inhibiting a signaling molecule or a signaling molecule's pathway, such as an inhibitor of SMAD signaling, inhibitor of glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ), refers to a compound or molecule (e.g., small molecule, peptide, peptidomimetic, natural compound, protein, siRNA, anti sense nucleic acid, aptamer, or antibody) that interferes with (i.e. reduces or suppresses or eliminates or blocks) the signaling function of the molecule or pathway.
  • an inhibitor is any compound or molecule that changes any activity of a named protein (signaling molecule, any molecule involved with the named signaling molecule, a named associated molecule, such as a glycogen synthase kinase 3 ⁇ (GSK3 ⁇ )) (e.g., including, but not limited to, the signaling molecules described herein), for one example, via directly contacting SMAD signaling, contacting SMAD mRNA, causing conformational changes of SMAD, decreasing SMAD protein levels, or interfering with SMAD interactions with signaling partners (e.g., including those described herein), and affecting the expression of SMAD target genes (e.g. those described herein).
  • a named protein signaling molecule, any molecule involved with the named signaling molecule, a named associated molecule, such as a glycogen synthase kinase 3 ⁇ (GSK3 ⁇ )
  • GSK3 ⁇ glycogen synthase kinase 3 ⁇
  • Inhibitors also include molecules that indirectly regulate SMAD biological activity by intercepting upstream signaling molecules.
  • an inhibitor of the present inventions induces (changes) or alters differentiation from a default to a non-default cell type, for example, one of the methods of the present inventions comprising LDN/SB, CHIR and a SHH activator (which may inhibit glycogen synthase kinase 3 ⁇ ) differentiated progenitor cells into non-default neural progenitor cells.
  • an inhibitor of the present invention s “alters” or “lowers” or “blocks” default signaling in order to direct cellular differentiation towards a nondefault cell type, such as described herein for differentiating floor plate midbrain progenitor cells and midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions.
  • an inhibitor of the present inventions is a natural compound or small molecule which changes signal molecule activity in a manner that contributes to differentiation of a starting cell population (day 0) into floor plate midbrain progenitor cells.
  • Inhibitors are described in terms of competitive inhibition (binds to the active site in a manner as to exclude or reduce the binding of another known binding compound) and allosteric inhibition (binds to a protein in a manner to change the protein conformation in a manner which interferes with binding of a compound to that protein's active site) in addition to inhibition induced by binding to and affecting a molecule upstream from the named signaling molecule that in turn causes inhibition of the named molecule.
  • an inhibitor is referred to as a “direct inhibitor” which refers to inhibiting a signaling target or a signaling target pathway by actually contacting the signaling target;
  • a direct inhibitor of a gamma secretase is a DAPT molecule that binds to the gamma secretase protein.
  • derivative refers to a chemical compound with a similar core structure.
  • floor plate midbrain progenitor cell in reference to an in vivo cell located in a midbrain, including during embryonic development of midbrain neurons, refers to a cell that may differentiate into a dopamine producing cell.
  • a “floor plate midbrain progenitor cell” refers to a cell in culture that is used to artificially produce a cultured cell in vitro that expresses overlapping or identical sets of markers when compared to markers expressed by in vivo cells, i.e.
  • a floor plate midbrain progenitor cell is “FOXA2+LMX1A+” or “FOXA2/LMX1A+”.
  • low numbers of cells in a differentiated progenitor population are FOXA2/LMX1A/TH+.
  • “authentic midbrain DA neurons” are FOXA2+/LMX1A+/NURR1+/TH+. These neurons were labeled “engraftable” after transplantation experiments in mice and primates showing the capability of these neurons to reverse Parkinson-like neurological conditions with less interference from neural overgrowth and teratoma formation.
  • the midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions were maintained in vitro for several months while retaining engrafting capability.
  • cells used for obtaining floor plate midbrain progenitor cells and midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons are obtained from a variety of sources including embryonic and nonembryonic sources, for example, hESCs and nonembryonic hiPSCs, somatic stem cells, disease stem cells, i.e. isolated pluripotent cells and engineered derived stem cells isolated from Parkinson disease patients, cancer stem cells, human or mammalian pluripotent cells, etc.
  • stem cell refers to a cell with the ability to divide for indefinite periods in culture and to give rise to specialized cells.
  • a stem cell may be obtained from animals and patients, including humans; for example, a human stem cell refers to a stem cell that is human.
  • a stem cell may be obtained from a variety of sources including embryonic and nonembryonic, such as umbilical cord cells, cells from children and cells from adults.
  • embryonic and nonembryonic such as umbilical cord cells, cells from children and cells from adults.
  • adult stem cells in general refer to cells that were not originally obtained from a fetus, in other words, cells from babies, cast off umbilical cords, cast off placental cells, cells from children, cells from adults, etc.
  • umbilical cord blood stem cells refer to stem cells collected from an umbilical cord at birth that have the capability to at least produce all of the blood cells in the body (hematopoietic).
  • the term “somatic (adult) stem cell” refers to a relatively rare undifferentiated cell found in many organs and differentiated tissues with a limited capacity for both self renewal (in the laboratory) and differentiation. Such cells vary in their differentiation capacity, but it is usually limited to cell types in the organ of origin.
  • the term “somatic cell” refers to any cell in the body other than gametes (egg or sperm); sometimes referred to as “adult” cells.
  • neural lineage cell refers to a cell that contributes to the nervous system (both central and peripheral) or neural crest cell fates during development or in the adult.
  • the nervous system includes the brain, spinal cord, and peripheral nervous system.
  • Neural crest cell fates include cranial, trunk, vagal, sacral, and cardiac, giving rise to mesectoderm, cranial cartilage, cranial bone, thymus, teeth, melanocytes, iris pigment cells, cranial ganglia, dorsal root ganglia, sympathetic/parasympathetic ganglia, endocrine cells, enteric nervous system, and portions of the heart.
  • adult stem cell refers to a somatic stem cell, for one example, a “hematopoietic stem cell” which refers to a stem cell in babies, children and adults, that gives rise to all red and white blood cells and platelets.
  • embryonic stem cell refers to a primitive (undifferentiated) cell that is derived from one of several sources, including but not limited to a preimplantation-stage embryo, an artificially created embryo, i.e. by in vitro fertilization, etc., capable of dividing without differentiating for a prolonged period in culture, and are known to have the capability to develop into cells and or tissues of the three primary germ layers, the ectoderm, the mesoderm, and the endoderm.
  • endoderm refers to a layer of the cells derived from the inner cell mass of the blastocyst; it has the capability to give rise to lungs, other respiratory structures, and digestive organs, or generally “the gut” “in vivo” and a variety of cell types in vitro.
  • embryonic stem cell line refers to a population of embryonic stem cells that have been cultured under in vitro conditions that allow proliferation without differentiation for up to days, months to years, for example, cells in a human WA-09 cell line.
  • human embryonic stem cell or “hESC” refers to a type of pluripotent stem cells derived from early stage human embryos, up to and including the blastocyst stage, that is capable of dividing without differentiating for a prolonged period in culture, and are known to develop into cells and tissues of the three primary germ layers, the ectoderm, the mesoderm, and the endoderm.
  • iPSC induced pluripotent stem cell
  • somatic (adult) cells are reprogrammed to enter an embryonic stem cell-like state by being forced to express factors important for maintaining the “stemness” of embryonic stem cells (ESCs).
  • ESCs embryonic stem cells
  • Mouse iPSCs demonstrate important characteristics of pluripotent stem cells, including the expression of stem cell markers, the formation of tumors containing cells from all three germ layers, and the ability to contribute to many different tissues when injected into mouse embryos at a very early stage in development.
  • Human iPSCs also express stem cell markers and are capable of generating cells characteristic of all three germ layers.
  • an iPSC is formed artificially by the introduction of certain embryonic genes (such as a OCT4, SOX2, and KLF4 transgenes) (see, for example, Takahashi and Yamanaka Cell 126, 663-676 (2006), herein incorporated by reference) into a somatic cell, for examples of cell lines from induced cells, C14, C72, and the like.
  • iPSC is an adult human skin cell, or fibroblast cell, transformed with using genes (OCT4, SOX2, NANOG, LIN28, and KLF4) cloned into a plasmid for example, see, Yu, et al., Science DOI: 10.1126/science.1172482, herein incorporated by reference.
  • totipotent refers to an ability to give rise to all cell types of the body plus all of the cell types that make up the extraembryonic tissues such as the placenta.
  • multipotent refers to an ability to develop into more than one cell type of the body.
  • pluripotent refers to a cell having the ability to give rise to at least two but often numerous different cell types of the body. Pluripotent cells often generate a teratoma after injection into an immunosuppressed mouse.
  • pluripotent stem cell refers to an ability of this cell to develop into at least two different cells types depending upon environmental factors, i.e. morphogens, growth factors, signaling molecules, either activators or inhibitors, etc.
  • a pluripotent stem cell refers to an ability of a cell to develop into any one of the three developmental germ layers of the organism including endoderm, mesoderm, and ectoderm.
  • specialized cell refers to a type of cell that performs a specific function in multicellular organisms.
  • groups of specialized cells such as neurons, work together to form a system, such as a nervous system.
  • neuroectoderm refers to a cell or cell fate found early in development or during pluripotent stem cell differentiation that can give rise to cells of the neural lineage.
  • markers of cell proliferation refers to the expression of molecules associated with rapidly cycling cells which are typically not present in mature slowly cycling or noncycling cells, i.e. actively dividing vs. cells with extended cycling times or noncycling cells.
  • markers include a Ki67 marker of cell proliferation (Gerdes, et al., Int J Cancer 31:13-20 (1983), herein incorporated by reference) and phospho-histone H3 markers of G2/M-phases of mitosis (Hendzel, et al., Chromosoma 106:348-360 (1997), herein incorporated by reference).
  • proliferation refers to an increase in cell number.
  • the term “differentiation” refers to a process whereby an unspecialized embryonic cell acquires the features of a specialized cell such as a specific type of neuron, brain cell, heart, liver, or muscle cell. Differentiation is controlled by the interaction of a cell's genes with the physical and chemical conditions outside the cell, usually through signaling pathways involving proteins embedded in the cell surface.
  • the term “differentiation” as used with respect to cells in a differentiating cell system refers to the process by which cells differentiate from one cell type (e.g., a multipotent, totipotent or pluripotent differentiable cell) to another cell type such as a target-differentiated cell.
  • one cell type e.g., a multipotent, totipotent or pluripotent differentiable cell
  • another cell type such as a target-differentiated cell.
  • cell differentiation refers to a pathway by which a less specialized cell (i.e. stem cell) develops or matures to possess a more distinct form and function (for example, an iPSC progressing into a neural crest progenitor to a cell of neuronal lineage to a floor plate midbrain progenitor cells to a midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions).
  • stem cell i.e. stem cell
  • DA dopamine
  • undifferentiated refers to a cell that has not yet developed into a specialized cell type.
  • default or “passive” in reference to a cell differentiation pathway refers to a pathway where a less specialized cell becomes a certain differentiated cell type in culture, when not treated with certain compounds i.e. normal cell cultures conditions without contact with at least one morphogen.
  • a default cell results when a cell is not contacted by a molecule capable of changing the differentiated cell type (i.e. a morphogen), for example cultures treated with LSB alone, but not an activator of SHH or an activator of Wnt for making a forkhead box protein A2 (FOXA2)+ cell of the present inventions, instead results in the expression of markers HES5, PAX6, LHX2, and EMX2.
  • non-default in reference to a cell refers to a differentiated cell type that results in a cell type that is different from a default cell, i.e. a non-default cell is a differentiated cell type resulting from a non-default conditions, such as cell of the present inventions, including a forkhead box protein A2 (FOXA2)+ neuronal cell, a floor plate midbrain progenitor cell and midbrain fate FOXA2/LMX1A+ dopamine (DA) neuron of the present inventions, etc.
  • FXA2 forkhead box protein A2
  • DA dopamine
  • a default cell may also be a default cell after a cell has contact with a morphogen to become a non-default cell without a subsequent morphogenic compound, such as a non-default floor plate midbrain progenitor cell that subsequently becomes a default cell that is not a midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons because of a lack of contact with a morphogen such as CHIR.
  • a morphogen such as CHIR.
  • morphogen refers to a compound that influences differentiation of a cell, i.e. determines, at least in part, cell fate. A morphogen also can influence a cell to differentiate into a non-default cell type.
  • the term “directed differentiation” refers to a manipulation of stem cell culture conditions to induce differentiation into a particular (for example, desired) cell type, such as floor plate midbrain progenitor cells and midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions.
  • the tam “directed differentiation” in reference to a cell refers to the use of small molecules, growth factor proteins, and other growth conditions to promote the transition of a cell from a pluripotent state into a more mature or specialized cell fate (e.g. central nervous system cell, neural cell, floor plate midbrain progenitor cell and midbrain fate FOXA2/LMX1A+ dopamine (DA) neuron of the present inventions, etc.).
  • the beginning of directed differentiation is the contacting of a cell at day 0 with LDN/SB.
  • a cell undergoing directed differentiation as described herein results in the formation of a non-default cell type of floor plate midbrain progenitor cells and midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions.
  • inducing differentiation in reference to a cell refers to changing the default cell type (genotype and/or phenotype) to a non-default cell type (genotype and/or phenotype).
  • inducing differentiation in a stem cell refers to inducing the cell to divide into progeny cells with characteristics that are different from the stem cell, such as genotype (i.e. change in gene expression as determined by genetic analysis such as a microarray) and/or phenotype (i.e.
  • a protein such as forkhead box protein A2 (FOXA2) or a set of proteins, such as forkhead box protein A2 (FOXA2) and LIM homeobox transcription factor 1, alpha (LMX1A) positive (+) while negative ( ⁇ ) for PAX6).
  • FOXA2 forkhead box protein A2
  • FOXA2 forkhead box protein A2
  • LIM homeobox transcription factor 1, alpha (LMX1A) positive (+) while negative ( ⁇ ) for PAX6 LIM homeobox transcription factor 1, alpha (LMX1A) positive (+) while negative ( ⁇ ) for PAX6
  • the term “fate” in reference to a cell in general refers to a cell with a genetically determined lineage whose progeny cells are capable of becoming a variety of cell types or a few specific cell types depending upon in vivo or in vitro culture conditions.
  • a cell's predetermined fate is determined by its environment to be destined for a particular differentiation pathway such that a cell becomes one cell type instead of another cell type, for example, a stem cell's progeny cells whose “neural fate” is to become a nerve cell instead of a muscle cell or a skin cell.
  • neural outgrowth or “neural outgrowth” refers to observation of elongated, membrane-enclosed protrusions of cytoplasm from cells.
  • neural overgrowth refers to unwanted unconstrained neural growth, i.e. uncontrolled growth of neurons, of transplanted cells at the site of engraftment.
  • teratoma refers to a noncancerous tumour from any tissue type growing from transplanted cells.
  • teratoma formation refers to the unwanted growth of a variety of tissue types into noncancerous tumours from growth of transplanted cells.
  • dopamine neuron or “dopaminergic neuron” in general refers to a cell capable of expressing dopamine.
  • “Midbrain dopamine neurons” or “mDA” refer to presumptive dopamine expressing cells in forebrain structures and dopamine expressing cells in forebrain structures.
  • neural stem cell refers to a stem cell found in adult neural tissue that can give rise to neurons and glial (supporting) cells. Examples of glial cells include astrocytes and oligodendrocytes.
  • floor plate or “FP” or “fp” refers to a region of the neural tube in vivo that extends along the entire ventral midline also described as the unpaired ventral longitudinal zone of the neural tube or referred to as a signaling center of the neural tube.
  • the neural tube was divided in different regions where the ventral cells closest to the midline constituted the floor plate.
  • chick midbrain FP can be divided into medial (MFP) and lateral (LFP) regions on the basis of gene expression, mode of induction and function.
  • MFP medial
  • LFP lateral
  • Floor plate cells are found in vivo in several areas of the developing embryo, for example floor plate cells in the midbrain, in the hindbrain, etc. In vivo, floor plate cells in the midbrain region are contemplated to give rise to cells that are different than cells differentiated from floor plate cells in other regions.
  • One primary floor plate marker in the midbrain region is FOXA2.
  • roof plate refers to the dorsal cells closest to the midline.
  • One roof plate marker is LMX1A.
  • floor plate and roof plate cells are located at distinct positions in the CNS (ventral versus dorsal) with diametrically opposed patterning requirements for their induction.
  • the term “midbrain” refers to a region of the developing vertebrate brain between the forebrain (anterior) and the hindbrain (posterior).
  • the midbrain regions gives rise to many areas of the brain, including but not limited to reticular formation, which is part of the tegmentum, a region of the brainstem that influences motor functions, the crus cerebri, which is made up of nerve fibers connecting the cerebral hemispheres to the cerebellum, and a large pigmented nucleus called the substantia nigra.
  • a unique feature of the developing midbrain is the co-expression of the floor plate marker FOXA2 and the roof plate marker LMX1A.
  • neuron refers to a nerve cell, the principal functional units of the nervous system.
  • a neuron consists of a cell body and its processes—an axon and one or more dendrites. Neurons transmit information to other neurons or cells by releasing neurotransmitters at synapses.
  • cell culture refers to a growth of cells in vitro in an artificial medium for research or medical treatment.
  • culture medium refers to a liquid that covers cells in a culture vessel, such as a Petri plate, a multiwell plate, and the like, and contains nutrients to nourish and support the cells. Culture medium may also include growth factors added to induce desired changes in the cells.
  • neuroneuronal maturation medium or “BAGCT” medium refers to a culture medium comprising N2 medium, further comprising brain-derived neurotrophic factor (BDNF), ascorbic acid (AA), glial cell line-derived neurotrophic factor, dibutyryl cAMP and transforming growth factor type ⁇ 3 for differentiating midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons.
  • BDNF brain-derived neurotrophic factor
  • AA ascorbic acid
  • glial cell line-derived neurotrophic factor glial cell line-derived neurotrophic factor
  • dibutyryl cAMP transforming growth factor type ⁇ 3 for differentiating midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons.
  • feeder layer refers to a cell used in co-culture to maintain pluripotent stem cells.
  • typical feeder layers include mouse embryonic fibroblasts (MEFs) or human embryonic fibroblasts that have been treated to prevent them from dividing in culture.
  • MEFs mouse embryonic fibroblasts
  • human embryonic fibroblasts that have been treated to prevent them from dividing in culture.
  • the term “passage” in reference to a cell culture refers to the process in which cells are disassociated, washed, and seeded into new culture vessels after a round of cell growth and proliferation. The number of passages a line of cultured cells has gone through is an indication of its age and expected stability.
  • the term “expressing” in relation to a gene or protein refers to making an mRNA or protein which can be observed using assays such as microarray assays, antibody staining assays, and the like.
  • paired box gene 6 or “PAX6” refers to a marker of a non-default neuroprogenitor cell.
  • TUJ1 or “neuron-specific class III beta-tubulin” in reference to a differentiating cell of the present inventions refers to a marker of early neural human cell differentiation, such as neural progenitor cells, and is found expressed in neurons of the PNS and CNS.
  • homodimer in reference to a SMAD molecule refers to at least two molecules of SMAD linked together, such as by disulfide linkages.
  • contacting cells with a compound of the present inventions refers to placing the compound in a location that will allow it to touch the cell in order to produce (obtain) “contacted” cells.
  • the contacting may be accomplished using any suitable method. For example, in one embodiment, contacting is by adding the compound to a tube of cells. Contacting may also be accomplished by adding the compound to a culture of the cells.
  • the term “attached cell” refers to a cell growing in vitro wherein the cell adheres to the bottom or side of the culture vessel, an attached cell may contact the vessel via extracellular matrix molecules and the like and requires the use of an enzyme for detaching this cell from the culture dish/container, i.e. trypsin, dispase, etc.
  • An “attached cell” is opposed to a cell in a suspension culture that is not attached and does not require the use of an enzyme for removing cells from the culture vessel.
  • markers refers to a gene or protein that identifies a particular cell or cell type.
  • a marker for a cell may not be limited to one marker, markers may refer to a “pattern” of markers such that a designated group of markers may identity a cell or cell type from another cell or cell type.
  • DA dopamine
  • midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions express one or more markers that distinguish a floor plate midbrain progenitor cell from a precursor less differentiated cell, i.e. forkhead box protein A2 (FOXA2) positive and LIM homeobox transcription factor 1, alpha (LMX1A) positive vs. HES5+ and PAX6+ cells, for example, as shown by exemplary gene expression patterns in FIGS. 1 e and 1 f.
  • the term “positive” in relation to a cell refers to a cell that expresses a marker, for one example, an antibody “stains” for that marker when using an antibody staining (detection) system or a nucleic acid sequence that hybridizes to the marker nucleic acid sequence as measured by a reporter molecule, i.e. a fluorescent molecule that attaches to double stranded nucleic acid sequences, in a detectable quantitative and/or qualitative amount above a control or comparative cell.
  • a reporter molecule i.e. a fluorescent molecule that attaches to double stranded nucleic acid sequences
  • a cell positive for a marker such as forkhead box protein A2 (FOXA2), etc.
  • FOXA2 forkhead box protein A2
  • Such as positive cell may be referred to as FOXA2+.
  • FOXA2+ When a cell is positive for more than one marker, such as when using the notation FOXA2/LMX1A+, the cell or the majority of the cell population is positive for both FOXA2 and LMX1A.
  • the term “negative” in relation to a cell or cell population, including a “negative cell” refers to a cell or population absent detectable signal for a marker or signal at levels of control populations.
  • a cell failing to stain following contacting with a forkhead box protein A2 (FOXA2) antibody detection method or gene array that includes detection of a FOXA2 mRNA, etc. is FOXA2- or negative for FOXA2.
  • FOXA2 forkhead box protein A2
  • reporter gene or “reporter construct” refer to genetic constructs comprising a nucleic acid encoding a protein that is easily detectable or easily assayable, such as a colored protein, fluorescent protein such as GFP or an enzyme such as ⁇ -galactosidase (lacZ gene).
  • GFP refers to any green fluorescent protein DNA sequence capable of producing a fluorescent protein upon expression in a cell typically used as an indication marker for expression of a target gene.
  • GFP include GFP sequences isolated from coelenterates, such as the Pacific jellyfish, Aequoria Victoria , and synthetic sequence derivatives thereof, such as “eGFP”.
  • sample is used in its broadest sense. In one sense it can refer to a cell or tissue. In another sense, it is meant to include a specimen or culture obtained from any source and encompasses fluids, solids and tissues.
  • Environmental samples include environmental material such as surface matter, soil, water, and industrial samples. These examples are not to be construed as limiting the sample types applicable to the present invention.
  • purified refers to the reduction in the amount of at least one contaminant from a sample.
  • a desired cell type is purified by at least a 10%, preferably by at least 30%, more preferably by at least 50%, yet more preferably by at least 75%, and most preferably by at least 90%, with a corresponding reduction in the amount of undesirable cell types, for example, directed differentiation of the present inventions resulted in the desired increase in purity of differentiated floor plate midbrain progenitor cells or midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons of the present inventions.
  • DA dopamine
  • purify refers to the removal of certain cells (e.g., undesirable cells) from a sample either mechanically, such as by flow cytometer cell sorting or through directed differentiation.
  • progenitor cells are purified by removal of contaminating PAX6 neuronal cells by sorting a mixed cell population into double positive forkhead box protein A2 (FOXA2)+ LIM homeobox transcription factor 1, alpha (LMX1A)+ cells by flow cytometry; midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons are also purified or “selected” from non-dopamine (DA) (default cells) by using a specified method of cell culture comprising compositions and methods of the present inventions.
  • non-midbrain fate FOXA2/LMX1A+ dopamine (DA) neuronal cells results in an increase in the percent of desired midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons in the sample.
  • purification of a cell type results in an “enrichment,” i.e., an increase in the amount, of the desired cell, i.e. midbrain fate FOXA2/LMX1A+ dopamine (DA) neurons in the sample.
  • Naturally occurring when applied to an object (such as cell, tissue, etc.) and/or chemical (such as a protein, amino acid sequence, nucleic acid sequence, codon, etc.) means that the object and/or compound are/were found in nature.
  • a naturally occurring cell refers to a cell that is present in an organism that can be isolated from a source in nature, such as an embryonic cell, wherein the cell has not been intentionally modified by man in the laboratory.
  • in vitro refers to an artificial environment and to processes or reactions that occur within an artificial environment.
  • in vitro environments exemplified, but are not limited to, test tubes and cell cultures.
  • in vivo refers to the natural environment (e.g., an animal or a cell) and to processes or reactions that occur within a natural environment, such as embryonic development, cell differentiation, neural tube formation, etc.
  • derived from or “established from” or “differentiated from” when made in reference to any cell disclosed herein refers to a cell that was obtained from (e.g., isolated, purified, etc.) a parent cell in a cell line, tissue (such as a dissociated embryo, or fluids using any manipulation, such as, without limitation, single cell isolation, cultured in vivo, treatment and/or mutagenesis.
  • a cell may derived from another cell, using for example chemical treatment, radiation, inducing new protein expression, for example, by infection with virus, transfection with DNA sequences, contacting (treating) with a morphogen, etc., and selection (such as by serial culture) of any cell type that is contained in cultured parent cells).
  • a derived cell can be selected from a mixed population by virtue of response to a growth factor, cytokine, selected progression of cytokine treatments, adhesiveness, lack of adhesiveness, sorting procedure, and the like.
  • the term “cell” refers to a single cell as well as to a population of (i.e., more than one) cells.
  • the population may be a pure population comprising one cell type, such as a population of neuronal cells or a population of undifferentiated embryonic cells.
  • the population may comprise more than one cell type, for example a mixed cell population. It is not meant to limit the number of cells in a population; for example, in one embodiment, a mixed population of cells may comprise at least one differentiated cell. In the present inventions, there is no limit on the number of cell types that a cell population may comprise.
  • the term “highly enriched population” refers to a population of cells, such as a population of cells in a culture dish, expressing a marker at a higher percentage or amount than a comparison population, for example, treating a LSB contacted cell culture on day 1 with purmorphamine and on day 3 with CHIR results in a highly enriched population of floor plate midbrain progenitor cells compare to treatment with LSB alone.
  • an enriched population is a population resulting from sorting or separating cells expressing one or more markers from cells not expressing the desired marker, such as a CD142 enriched population, an A9 enriched population, and the like.
  • cell biology refers to the study of a live cell, such as anatomy and function of a cell, for example, a cell's physiological properties, structure, organelles, and interactions with their environment, their life cycle, division and death.
  • nucleotide sequence of interest refers to any nucleotide sequence (e.g., RNA or DNA), the manipulation of which may be deemed desirable for any reason (e.g., treat disease, confer improved qualities, expression of a protein of interest in a host cell, expression of a ribozyme, etc.), by one of ordinary skill in the art.
  • nucleotide sequences include, but are not limited to, coding sequences of structural genes (e.g., reporter genes, selection marker genes, oncogenes, drug resistance genes, growth factors, etc.), and non-coding regulatory sequences which do not encode an mRNA or protein product (e.g., promoter sequence, polyadenylation sequence, termination sequence, enhancer sequence, etc.).
  • protein of interest refers to a protein encoded by a nucleic acid of interest.
  • the term “gene” refers to a nucleic acid (e.g., DNA or RNA) sequence that comprises coding sequences necessary for the production of a polypeptide or precursor (e.g., proinsulin).
  • the polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence so long as the desired activity or functional properties (e.g., enzymatic activity, ligand binding, signal transduction, etc.) of the full-length or fragment are retained.
  • the term also encompasses the coding region of a structural gene and includes sequences located adjacent to the coding region on both the 5′ and 3′ ends for a distance of about 1 kb or more on either end such that the gene corresponds to the length of the full-length mRNA.
  • sequences that are located 5′ of the coding region and which are present on the mRNA are referred to as 5′ untranslated sequences.
  • sequences that are located 3′ or downstream of the coding region and which are present on the mRNA are referred to as 3′ untranslated sequences.
  • the term “gene” encompasses both cDNA and genomic forms of a gene.
  • a genomic form or clone of a gene contains the coding region interrupted with non-coding sequences termed “introns” or “intervening regions” or “intervening sequences.” Introns are segments of a gene that are transcribed into nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers.
  • Introns are removed or “spliced out” from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (mRNA) transcript.
  • mRNA messenger RNA
  • the mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide.
  • RNA expression refers to the process of converting genetic information encoded in a gene into RNA (e.g., mRNA, rRNA, tRNA, or snRNA) through “transcription” of the gene (i.e., via the enzymatic action of an RNA polymerase), and for protein encoding genes, into protein through “translation” of mRNA.
  • Gene expression can be regulated at many stages in the process. “Up-regulation” or “activation” refers to regulation that increases the production of gene expression products (i.e., RNA or protein), while “down-regulation” or “repression” refers to regulation that decrease production. Molecules (e.g., transcription factors) that are involved in up-regulation or down-regulation are often called “activators” and “repressors,” respectively.
  • nucleic acid molecule encoding refers to the order or sequence of deoxyribonucleotides or ribonucleotides along a strand of deoxyribonucleic acid or ribonucleic acid. The order of these deoxyribonucleotides or ribonucleotides determines the order of amino acids along the polypeptide (protein) chain. The DNA or RNA sequence thus codes for the amino acid sequence.
  • isolated when used in relation to a nucleic acid, as in “an isolated oligonucleotide” or “isolated polynucleotide” refers to a nucleic acid sequence that is identified and separated from at least one component or contaminant with which it is ordinarily associated in its natural source. Isolated nucleic acid is such present in a form or setting that is different from that in which it is found in nature. In contrast, non-isolated nucleic acids as nucleic acids such as DNA and RNA found in the state they exist in nature.
  • a given DNA sequence e.g., a gene
  • RNA sequences such as a specific mRNA sequence encoding a specific protein
  • isolated nucleic acid encoding a given protein includes, by way of example, such nucleic acid in cells ordinarily expressing the given protein where the nucleic acid is in a chromosomal location different from that of natural cells, or is otherwise flanked by a different nucleic acid sequence than that found in nature.
  • the isolated nucleic acid, oligonucleotide, or polynucleotide may be present in single-stranded or double-stranded form.
  • the oligonucleotide or polynucleotide will contain at a minimum the sense or coding strand (i.e., the oligonucleotide or polynucleotide may be single-stranded), but may contain both the sense and anti-sense strands (i.e., the oligonucleotide or polynucleotide may be double-stranded).
  • kit refers to any delivery system for delivering materials.
  • a kit may refer to a combination of materials for contacting stem cells, such delivery systems include systems that allow for the storage, transport, or delivery of reaction reagents from one location to another in the appropriate containers (such as tubes, etc.) and/or supporting materials (e.g., buffers, written instructions for performing cell differentiation, etc.) (e.g., compounds, proteins, detection agents (such as antibodies that bind to tyrosine hydroxylase (TH), forkhead box protein A2 (FOXA2), LIM homeobox transcription factor 1, alpha (LMX1A), etc.), etc.
  • TH tyrosine hydroxylase
  • FOXA2 forkhead box protein A2
  • LIM homeobox transcription factor 1, alpha (LMX1A) LIM homeobox transcription factor 1, alpha
  • kits include one or more enclosures (e.g., boxes, or bags, test tubes, Eppendorf tubes, capillary tubes, multiwell plates, and the like) containing relevant reaction reagents for inhibiting signaling pathways, for example, an inhibitor for lowering transforming growth factor beta (TGF ⁇ )/Activin-Nodal signaling, such as SB431542 (or SB431542 replacement), and the like, an inhibitor for lowering SMAD signaling, LDN-193189 (or LDN-193189 replacement), and the like, an inhibitor for lowering glycogen synthase kinase 3 ⁇ (GSK3 ⁇ ), for one example, for activation of wingless (Wnt or Wnts) signaling otherwise known as a WNT signaling activator (WNT agonist), such as CHIR99021 (or CHIR99021 replacement), etc.), and the like, an activator of Sonic hedgehog (SHH) signaling (such as a smoothened (SMO) receptor small molecule agonist), for example, a Sonic hedgehog (
  • the reagents in the kit in one embodiment may be in solution, may be frozen, or may be lyophilized.
  • the reagents in the kit in one embodiment may be in individual containers or provided as specific combinations, such as a combination of LSB (LDN-193189 with SB431542), Sonic hedgehog (SHH) C25II molecule with purmorphamine, Sonic hedgehog (SHH) C25II molecule with purmorphamine with CHIR99021 or purmorphamine with CHIR99021, neuronal maturation molecules and the like.
  • FIG. 1 shows exemplary induction and neurogenic conversion of human ES cell-derived midbrain floor plate precursors dependent on CHIR990221 addition.
  • e,f Lists of selected differentially expressed genes at day 11 comparing LSB/S/F8/CHIR conditions with either LSB (e) or LSB/S/F8 (f).
  • g,h Temporal gene expression analysis of selected markers characteristic of midbrain DA precursor identity (g), forebrain and ventral non-DA precursor identity (h). Scale bars correspond to 50 ⁇ m.
  • FIG. 2 shows an exemplary immunocytochemical and molecular analysis of midbrain DA neuron fate in LSB/S/F8/CHIR treated versus LSB/S/F8 (ventral/hypothalamic) and LSB (dorsal forebrain) fates.
  • c,d Global gene expression analysis was performed at day 25 (triplicate samples for all three conditions). Selected lists of the most differentially expressed genes comparing day 13 versus day 25 in the LSB/S/F8/CHIR condition (c) and comparing LSB/S/F8/CHIR treatment versus LSB (d, left panel) and LSB/S/F8 (d, right panel). e) Normalized differential gene expression analysis for key midbrain DA neuron markers. Significance levels for individual markers are presented as compared to LSB only treatment: ANOVA; Dunnett test: *** p ⁇ 0.001; ** p ⁇ 0.01; p ⁇ 0.05). Scale bars correspond to 50 ⁇ m.
  • FIGS. 3-1 and 3-2 shows an exemplary in vitro maturation, characterization and functional assessment of floor plate derived- versus rosette-derived midbrain DA neurons.
  • FIG. 3-1 shows exemplary: a) Immunocytochemical analysis at day 50 of differentiation for TH (red), in combination with LMX1A (green, left panels), FOXA2 (blue, left panels) and NURR1 (green, right panels). b) Quantification of TH+, FOXA2+, LMX1+ and NURR1+ cells out of total cells in rosette-derived versus floor plate-derived (LSB/S/F8/CHIR) cultures.
  • FIG. 3-2 shows an exemplary summary of cells produced by a floor plate based midbrain DA neuron protocol as described herein. a) In contrast to past strategies (for example, Perrier, A. L. et al. Derivation of midbrain dopamine neurons from human embryonic stem cells.
  • FIG. 4 shows an exemplary in vivo survival and function of floor plate-derived human DA neurons in mouse, rat and monkey PD model host brain.
  • a-d Transplantation of floor plate-derived DA neurons in 6-OHDA lesioned adult mice (NOD-SCID IL2Rgc null strain).
  • c) Quantification of FOXA2+, TH+ and double-labeled cells in floor plate-derived grafts (mean ⁇ SEM, n 4 at 4.5 months post grafting).
  • h-j High power images showing co-expression of TH (green) with midbrain specific transcription factors FOXA2, PITX3 and NURR1 (red).
  • k-m Behavioral analysis of animals treated with floor plate-derived DA neuron grafts versus sham-treated animals.
  • k Amphetamine-induced rotational asymmetry.
  • l stepping test: measuring forelimb akinesia in affected versus non-affected side.
  • n-p Immunohistochemical analysis for TH (green) and co-expression (red) with DAT (n), GIRK2 (o) and calbindin (p). Significance levels (panels d, k, l, m) are: ** p ⁇ 0.01; p ⁇ 0.05). Scale bars correspond to 200 ⁇ m in (e), 50 ⁇ m in (f), 20 ⁇ m in (h-j) and 40 ⁇ m in (n-p).
  • q-t Transplantation of floor plate-derived DA neurons into adult 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) lesioned rhesus monkey.
  • FIG. 5 shows an exemplary timing of CHIR99021 exposure determines induction of FOXA2/LMX1A midbrain floor plate precursors.
  • FIG. 6 shows an exemplary FGF8 exposure does not play a major role in the induction of FOXA2/LMX1A midbrain floor plate precursors.
  • FIG. 7 shows an exemplary exposure to high dose of SHH and/or a smoothened small molecule agonist (purmorphamine) is required for efficient midbrain floor plate induction in the presence of CHIR99021.
  • FIG. 8 shows an exemplary analysis of genes differentially expressed in LSB/S/F8/CHIR treated versus LSB and LSB/S/F8 treated cultures at days 11 and 25 of differentiation.
  • FIG. 9 shows an exemplary differentiation protocol for floor plate induction and midbrain DA neuron development in representative independent hESC and hiPSC lines.
  • Data from the hESC line H1 and the hiPSC lines 2C6 (from a sporadic PD patient) and SeV6 (Sendai-based, integration-free) are presented.
  • the floor plate based protocol described in FIG. 1 d and FIG. 10 was used followed by analysis of FOXA2 (red) expression at day 11 and TH (green)/FOXA2 (red) at day 25 of differentiation.
  • FIG. 10 shows an exemplary schematic summary of the differentiation conditions used for floor plate-derived and rosette-derived DA neuron cultures. Both protocols used dual-SMAD inhibition to accelerate neural fate acquisition. LDN was used for BMP inhibition in the floor plate protocol while the traditional noggin induction was used for rosette cultures. The abbreviations are: LDN: LDN-193189, SB: SB431542, SHH (purmorphamine+SHH C25II), FGF8: FGF8, BAGCT: BDNF+ascorbic acid+GDNF+dbcAMP+TGF ⁇ 3. SHH/FGF8 in the rosette protocol used SHH C25I alone in the absence of purmorphamine following the initial recommendation for patterning of rosette-derived DA neuron cultures. Note: Purmorphamine treatment at rosette stage shows toxicity at concentration suitable for patterning floor plate cells. BASF: BDNF+ascorbic acid+SHH/FGF8.
  • FIG. 11 shows an exemplary in vitro maturation of floor plate-derived DA neuron cultures.
  • Scale bars correspond to 20 ⁇ m in (a) 100 ⁇ m in (b) and 20 ⁇ m in (c).
  • FIG. 13 shows an exemplary histological analysis of long-term (4.5 months) grafted 6-OHDA lesioned mice (NOD-SCID IL2Rgc null strain) comparing behavior of floor plate- versus rosette-derived grafts. Analysis of floor plate-derived grafts.
  • b) Robust hNCAM+ fiber outgrowth was observed at graft periphery.
  • Serotonergic (5-HT+) fibers (green) in graft are largely negative for hNCAM (red) suggesting host origin.
  • d GABAergic neurons and fibers (green) in graft. Analysis of rosette-derived grafts: e) Neural overgrowth with compression of host brain tissue. The majority of cells were positive for both NCAM (red) and DCX (green) suggesting neuronal fate. f) NCAM+/DCX+ fibers extended to the non-transplanted contra-lateral side of the brain. g) Within the graft core multiple DCX+ clusters were observed. h) Few TH+ cells (green) and fibers were observed at graft periphery and nearly all rosette-derived TH+ cells in vivo were negative for FOXA2 (blue).
  • FIG. 14 shows an exemplary histological analysis of long-term (5 months) grafted 6-OHDA lesioned SD rats.
  • GFAP+ fibers (green) within the graft did not co-express hNA (red) suggesting host origin.
  • FIG. 15 shows an exemplary histological analysis of floor plate-derived grafts in primate brain.
  • a) Upper left and right panels: High-resolution image reconstructions of representative graft sites (1 month after transplantation). Graft cytoarchitecture is illustrated by immunohistochemistry for the human specific marker SC-121 (no cross-reactivity with non-human primate tissues).
  • Lower left panel SC-121+ fibers extending from graft core.
  • Lower right panel Higher resolution image of SC-121+ cells showing neuronal precursor morphologies.
  • Scale bars correspond to 500 ⁇ m in (a, top panel), 50 ⁇ m in (a, lower left panel), 20 ⁇ m in (a, lower right panel), 2 mm in (b), 500 ⁇ m in (c, left panel), 50 ⁇ m (c, upper right panel), 100 ⁇ m (c, lower right panel), 100 ⁇ m (d).
  • FIG. 16 shows an exemplary derivation of TH+ cells from hESCs using a previous MS5 feeder cell based method that differentiated cells into DA neuron-like cells via (through) rosette cell intermediates.
  • Top at P0 hESCs were contacted with molecules for beginning neural induction of Oct4+ cells into rosette cells using MS5 feeder cells (Perrier et al., 2004).
  • At the P1 stage rosette cells were expanded by contacting cells with additional molecules for differentiating cells into cells at stage P2 with specific expression patterns including Pax2+/En1+ DA progenitor cells further differentiated into TH+/En1+ DA neurons.
  • FIG. 16C few graft-derived cells (hNA+ (green) co-express TH (red) suggesting that most grafted human cells adopt a non-DA neuron phenotype.
  • Panels 16 D-E show that D-E, despite the very poor in vivo survival there was some (albeit very modest and highly variable) improvement in a few behavioral assays such as amphetamine induced rotations (D), cylinder test and spontaneous rotations (E).
  • FIG. 17 shows an exemplary protocol for derivation of low numbers of floor plate cells.
  • a modified Dual-SMAD inhibition protocol generated floor plate (fp) cells. High concentrations of SHH were necessary for the induction of FoxA2+ fp cells and that addition of caudalizing patterning cues such as FGF8, Wnt1 or RA did not lead to decrease in FOXA2 expression but change in regional identity
  • FoxA2+ floor plate cells are only induced in the presence of high dose SHH. Addition of RA. Wnt1 and FGF8 does not inhibit FoxA2 induction.
  • FIG. 18 shows an exemplary protocol for derivation of floor plate cells showing high levels of midbrain characteristics as compared to the low or absent levels in cells made from the procedures used in FIGS. 16 and 17 .
  • Chart B shows specifically the genes that are common among the three treatment conditions versus those genes that are unique to each individual condition.
  • Chart B is a preliminary analysis used for the microarray results presented in FIG. 1 .
  • C mRNA levels of midbrain markers FoxA2, LMX1A, are highly enriched in LSB/SHH/FGF8/CHIR-treated group compared to SHH/FGF-treated group.
  • FIG. 19 shows an exemplary in vitro characterization of dopamine neurons derived from the midbrain region of the floor plate.
  • A Co-labeling of FoxA2+ neurons with mDA neuron markers TH, Nurr1 and LMX1A at d25 of differentiation.
  • B mRNA expression levels by QRT-PCR of mDA neuron markers as well as other midbrain cell types in LDN/SB+SHH/FGF8+SHH/FGF8+CHIR treated groups.
  • FIG. 20 shows an exemplary comparable differentiation potential towards midbrain DA neuron fate of PINK1 mutant PD-iPS cells versus wild-type hES (or iPS) cells.
  • PINK1 Q456X mutant PD-iPSC line was differentiated using the novel floor-plate based midbrain DA neurons methods of the present inventions yielding midbrain differentiation profiles comparable to those obtained from H9 line.
  • A-C Immunocytochemical analysis of PINK1 mutant PD-iPSC line at day 11 of differentiation (midbrain precursor stage) for FOXA2 (red), LMX1A (green) and DAPI (blue) (A), day 25 of differentiation (early postmitotic DA neuronal stage) for FOXA2 (red) and TH (green) (B) and for NURR1 (red) and TH (green) (C).
  • FIG. 21 shows an exemplary PINK1 mutant PD-iPSC showed PD like phenotype of protein aggregation following long-term differentiation and maturation in vitro.
  • PINK1 mutant PD-iPSC showed evidence of ⁇ -synuclein (a major component of Lewy body formation in PD patients) expression in cytosol of TH+ DA neurons.
  • the cells also showed high expression of ubiquitin (a classical Lewy body marker).
  • DA neurons derived from control iPS line showed expression of normal synaptic (as opposed to cytosolic) ⁇ -synuclein expression and very low levels of ubiquitin.
  • FIG. 22 shows an exemplary expression of aggregated form of ⁇ -synuclein.
  • dimerized insoluble forms of a-synuclein lead to aggregation in Lewy body.
  • the dimerized form of ⁇ -synuclein shows phospholylation of Serine 129 on ⁇ -synuclein.
  • PINK1 mutant PD-iPSC derived cells showed strong expression for Ser129 phosphorylated ⁇ -synuclein in contrast to control-iPSC derived cells that showed very low levels of expression.
  • FIG. 23 shows exemplary differences in ⁇ -synuclein expression patterns are observed depending of differentiation protocol.
  • the inventors' show that ‘authentic’ midbrain DA neurons have PD specific vulnerability and corresponding, specific in vitro phenotypes.
  • DA neurons obtained using the classical MS5 stromal feeder based differentiation protocol can yield large numbers of TH+ neurons.
  • the TH+ cells resulting from differentiation by the classical MS5 stromal feeder protocol are not authentic midbrain DA neurons.
  • cultures differentiated via the MS5 protocol there were many ⁇ -synuclein positive cells. However, those cells did not co-express TH.
  • A B) Immunocytochemical analysis for ⁇ -synuclein (LB509, red), TH (green) of PINK1 mutant PD-iPSC line at day 60 of MS5 based differentiation (A) and control-iPSC (B).
  • FIG. 24 shows an exemplary DA neurons derived from PINK1 mutant PD-iPSC are more vulnerable to toxic stimulation.
  • PD-iPSC derived TH+ DA neurons derived via the floor-plate based protocols of the present inventions were more vulnerable to toxin challenge (valinomycin: mitochondria ionophore, 5 uM, 48 hr) than control-iPSC derived cells.
  • valinomycin mitochondria ionophore, 5 uM, 48 hr
  • TH+ neurons derived via the classic MS5 based protocol did not show differential vulnerability between PD- versus control-derived cells.
  • A-F Representative TH immunocytochemistry at day 60 of differentiation: Normal condition (no toxin treatment) for both PD- and control-iPSC derived cells obtained via floor-plate based protocol (A, PD-iPSC derived cells shown), nearly complete degeneration of TH+ DA neurons in PD-iPSC following toxin treatment (B), partially degenerated TH+ DA neurons from control-iPSC(C), Normal condition both of PD- and control-iPSC derived cultures obtained via MS5 based protocol (D, PD-iPSC derived cells shown), TH+ neurons following toxin challenge in PD-iPSC (E), and control-iPSC derived cultures (F) obtained via MS5 protocol.
  • G-H low power images of immunocytochemistry for Tuj1 (red) and TH (green) by floor-plate based protocol at day 60 of differentiation: PD-iPSC of normal (G), versus toxin challenge (H) conditions and control iPSC of normal (I), versus toxin challenge (J) conditions.
  • K-N low power images of immunocytochemistry for Tuj1 (red) and TH (green) by MS5 based protocol at day 60 of differentiation: PD-iPSC of normal (K), versus toxin challenge (L) conditions and control iPSC of normal (M), versus toxin challenge (N) conditions.
  • FIG. 25 shows an exemplary quantification of cell viability-dose response assay for toxin challenge.
  • Cell viability assay with alamar-blue after 48 hrs of valinomycin treatment showed differential cell survival in a specific dose range for toxin challenge (5 and 10 uM) when comparing PD-iPSC and control iPSC (day 60 of floor-plate based differentiation of the present inventions). Note: this assay tests for overall cell death while the most dramatic effects were observed specifically in DA neurons (see FIG. 14 ). Therefore, alamar blue based quantification will likely underestimate the extent of the differential effect observed on DA neuron lineages.
  • FIG. 26 shows exemplary grafted human DA neurons derived from pluripotent stem cells have electrophysiological features typical of those seen in mouse substantia nigra pars compacta (SNpc).
  • B Top-cell-attached patch recording from a putative DA neuron in the graft; Bottom-whole cell recording from the same cell.
  • FIG. 27 shows an exemplary A9 candidate surface marker and CD-screen in hPSCs.
  • a) Venn Diagram of transcriptome data from FACS purified mouse ESC derived mDA neurons. Among the 107 genes shared between PITX3 and Nurr1 the majority were known markers of midbrain DA neurons as well as novel markers were confirmed expressed within the ventral midbrain: b) One of those markers was DCSM1, a putative surface marker that appears to be enriched within the A9 region, based on mRNA in situ expression data (Allen Brain Atlas, Lein, E. S. et al., Genome-wide atlas of gene expression in the adult mouse brain, Nature 445: 168-176 (2007)).
  • CD screen c) representative 96 well plate (1 out of 3 ⁇ 96 plates used to screen complete CD panel). Dark wells label CD markers that are highly expressed in hESC DA neurons at day 25. e) Summary of the CD screening results in hESC derived DA neurons. f) One exemplary marker, CD142, a surface marker enriching specifically for DA neurons at the Nurr1+ stage was as found following FACS mediated isolation of CD142+ versus CD142 ⁇ cells and analysis at day 7 post sort.
  • FIG. 28 shows exemplary CD142 enriched for Nurr1+ midbrain DA neuron stage and depletes for GABA and Serotonergic neurons.
  • CD142 depletes GABAergic and Serotonergic contaminants following day 25 sorting and in vitro culture for 3 weeks.
  • FIG. 29 shows an exemplary contemplated experimental use of PSA.
  • PST-expressing and PSTnm exposed hESC derived DA neurons will be assessed in vitro for impact on DA phenotype and fiber outgrowth.
  • In vivo studies in 6OHDA rat model will be tested for whether lower numbers of DA neurons with forced PSA expression can match behavioral recovery of standard grafts, and whether forced PSA expression in hESC-derived DA neurons is capable of inducing recover in assays of complex motor function.
  • FIG. 30 shows an exemplary use of PST.
  • Overexpression mouse PST
  • A Quantification of PST mRNA by qPCR in control cells (Nurr1) and in cells overexpressing PST (Nurr1/PST). Data is expressed as the fold enrichment of PST levels in Nurr1/PST versus Nurr1 cells.
  • B PSA immunostaining in DA neuron cultures at day 14 of differentiation shows increased levels of PSA in Nurr1/PST cells (Scale bar: 100 ⁇ m).
  • C Western Blot for NCAM in differentiated cells.
  • Nurr1/PST cells shows increased levels of the polysialylated form of NCAM (smear, brackets) compared to control (lane 1). PSA is removed from NCAM after endoN treatment (lane 3).
  • D Quantification of the intensity of the PSA smear expressed in arbitrary units.
  • E PSA FACS analysis at day 14 of differentiation. Treatment of cells with 20 units of endoN, 24 hours before the end of differentiation, abolished the PST effect.
  • F Representative photomicrographs comparing Nurr1 and Nurr1/PST differentiated cells for GFP immunofluorescence and DA markers. Cells sorted for GFP and re-plated still retained the DA phenotype (post sort). Scale bars: 100 ⁇ m.
  • FIG. 31 shows an exemplary FACS analysis of ES-derived DA neurons.
  • Flow cytometry-based isolation of GFP+ and SSEA-1 ⁇ cells As double negative and GFP negative controls, J1 mouse ES-cells were used. Around 5 to 10% of cells were sorted positively.
  • FIG. 32 shows exemplary Nurr1/PST grafts were more effective at inducing behavioral recovery in 6OHDA mouse model.
  • Nurr1::GFP cells were differentiated and sorted at day 14 for GFP+/SSEA-1 ⁇ population.
  • Cells treated with endoN were cultured for 12 hours before sorting with 20 units of the enzyme. 55,000 cells were grafted in 1 ml of N2 media with BDNF and AA.
  • B There were more GFP+ cells in the PST graft at endpoint than in control (p ⁇ 0.05, t-test).
  • D GFP, TH and PSA immunofluorescence. Scale bar: 200 ⁇ m.
  • E Grafted cells express DA markers. Individual z-planes of confocal micrographs are shown. Scale bar: 20 ⁇ m. Values are means+/ ⁇ SEM.
  • FIG. 33 shows exemplary PSA augmentation that increased host striatum innervation by ES-derived DA neurons.
  • PSA-NCAM overexpression increased process outgrowth.
  • A Representative photomicrographs of GFP/TH+, GFP/Girk2+ and GFP/synapsin+ processes in controls. Staining in Nurr1/PST samples was similar. Scale bar: 20 ⁇ m.
  • B Representative z-stack projections showing GFP+ processes extending out of both Nurr1 and Nurr1/PST grafts. There are more GFP+ processes extending out of the Nurr1/PST graft (scale bar: 50 ⁇ m). Insets show GFP+/TH+ processes in same sections. Arrow: direction of growth.
  • FIG. 34 shows an exemplary use of PST in methods associated with spinal cord injury and for expression on motoneurons.
  • SCs grafted GFP Schwann cells
  • A host scar tissue
  • B PST-modified SCs readily migrate considerable distances
  • This PSA engineering resulted in improvement in locomotion, BBB subscore; upper line in (C) shown vs lower line control) and hindlimb dexterity (gridwalk test; lower line in (D) vs upper line controls).
  • FIG. 35 shows an exemplary use of PSTnm enzyme.
  • PSTnm-produced PSA inhibits adhesion of Schwann cells in suspension to a Schwann cell monolayer even more effectively (red line-lowest line) than PSA produced by forced PST expression (green line-middle line).
  • B PSA immunoblotting in ESC-derived HB9 motoneurons shows that control samples treated with PSTnm have undetectable levels of PSA. Incubation with PSTnm+CMP-sialic acid substrate produces a large PSA band, which is removed with endoN treatment.
  • the present invention relates to the field of stem cell biology, in particular the lineage specific differentiation of pluripotent or multipotent stem cells, which can include, but is not limited to, human embryonic stem cells (hESC) in addition to nonembryonic human induced pluripotent stem cells (hiPSC), somatic stem cells, stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • hESC human embryonic stem cells
  • hiPSC human induced pluripotent stem cells
  • somatic stem cells stem cells from patients with a disease, or any other cell capable of lineage specific differentiation.
  • DA dopamine
  • the midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons made using the methods of the present invention are further contemplated for various uses including, but not limited to, use in in vitro drug discovery assays, neurology research, and as a therapeutic to reverse disease of, or damage to, a lack of dopamine neurons in a patient. Further, compositions and methods are provided for differentiating midbrain fate FOXA2+LMX1A+TH+ dopamine (DA) neurons from human pluripotent stem cells for use in disease modeling, in particular Parkinson's disease.
  • the present inventions relate to characteristics of Parkinson's disease (PD) including the selective degeneration of midbrain dopamine (mDA) neurons in patients' brains.
  • PD symptoms are primarily due to the selective loss of DA neurons in the substantia nigra of the ventral midbrain, PD is considered one of the diseases most suitable for cell replacement therapeutic strategies for treatment.
  • mDA midbrain dopamine
  • hPSCs Human pluripotent stem cells
  • spinal motoneurons Li, et al. Nat. Biotechnol. 23, 215-221 (2005), herein incorporated by reference
  • DA midbrain dopamine
  • dopamine (DA)-like neurons are not authentic midbrain dopamine (DA) neurons of the present inventions (see FIGS. 3, 10, 13 and 16 ). Therefore, the inventors labeled the floor-plate derived dopamine producing neurons made by methods described herein, i.e. dopamine producing neurons of the present inventions as “authentic” because unlike dopamine producing neurons made by published methods, when “authentic” dopamine producing neurons of the present inventions are transplanted into rodents and primates they reverse Parkinson-like neurological conditions with less interference from neural overgrowth and teratoma formation. Also, unlike previous methods of making dopamine producing neurons, “authentic” dopamine producing neurons of the present inventions are produced at higher percentages from starting populations and retain engrafting capability for several months in culture.
  • hPSC derived neurons are used for transplantation and are related to their potential for teratoma formation or neural overgrowth (Roy, et al. Nature Med. 12, 1259-1268 (2006); Elkabetz, et al. Genes Dev. 22, 152-165 (2008), herein incorporated by reference).
  • DA neurons derived from human ESCs Another possible source of cells for transplantation are DA neurons derived from human ESCs. Previous attempts using these cells as a starting cell population to make differentiated cells that appeared to be DA like-midbrain neurons derived from human embryonic stem cells (hESCs) that were transplanted into rodent PD models resulted in poor in vivo survival of the transplants after transplantation. This failure was contemplated to most likely be due to incomplete midbrain DA neuron differentiation in vitro resulting in cells that appeared to be midbrain DA neurons but were not capable of engraftment to replace lost neuron function.
  • hESCs human embryonic stem cells
  • DA-like neurons previously made in their laboratories and described in publications were not the same cell type nor had similar functions or engraftment capabilities as the floor-plate midbrain DA neuronal cells of the present inventions, see, FIGS. 16 and 17 for examples. Therefore the inventors also discovered that in order for cells to undergo directed differentiation in the laboratory to produce cell populations containing large numbers of properly functioning neurons, the cells needed to go through the specific developmental stages in order to become a suitable replacement cell population for cell based replacement therapies. The inventors also discovered that, at least for obtaining the engraftable DA neurons of the present inventions, certain developmental stages must be present, such as the FOX2A/LIM1A+ Day 11 intermediates. If such developmental stages are not present, the inventors' discovered that resulting DA-like neurons do not have the same functional capabilities as the midbrain DA neurons of the present inventions that were derived from FOX2A/LIM1A+ Day 11 intermediates.
  • midbrain FOXA2+/LMX1A+ floor plate precursors were derived from hPSCs in 11 days following exposure to small molecule activators of sonic hedgehog (SHH) and canonical WNT signaling. These Day 11 cells, double positive for FOXA2+ and LMX1A+, are contacted with additional small molecules to induce further differentiation into engraftable midbrain DA neurons, positive for TH+FOXA2+ and LMX1A+, by day 25. These mature floor-plate midbrain DA neurons can be maintained in vitro for several months. Extensive in vitro molecular profiling, biochemical and electrophysiological data defined developmental progression and confirmed identity of hPSC-derived midbrain DA neurons.
  • hESC human embryonic stem cell
  • hiPSCs human induced pluripotent stem cells
  • hESC or hiPSC Human stem cells populations
  • hESC or hiPSC Human stem cells populations
  • This protocol was used to demonstrate high yields of hESC progeny by Day 11 of directed differentiation into a midbrain DA (mDA) neuronal phenotype which included expression of key transcription factors e.g. TH, FoxA2 and LMX1A which upon further differentiation yielded additional key proteins e.g. TH.
  • mDA midbrain DA
  • human ES cells were differentiated into early neuroepithelial cells, termed neural rosettes, followed by induction of rosette stage cells into cells expressing midbrain DA neuron precursor and differentiated markers. Those cells went on to exhibit functional neuronal features by electrophysiology, in vitro DA release and the formation of TH-immunogold positive synaptic contacts (Perrier, et al. From the Cover: Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci USA 101, 12543-8 (2004), herein incorporated by reference).
  • transplantation of the cells in 6OHDA lesioned murine hosts resulted in a very small number of surviving dopaminergic neurons.
  • the following example describes exemplary methods for providing cells of a neural lineage for use during development of the present inventions.
  • Dual SMAD inhibition was previously used as a rapid and highly effective method for inducing one type of neural lineage cells from hPSCs (Chambers, et al., Nat Biotechnol 27, (2009), herein incorporated by reference).
  • These neural lineage cells induced by molecules, including Noggin had a default pathway that allowed development into central nervous system cells, i.e. neural cell fate.
  • DM dorsomorphin
  • LDN was used in place of Noggin to inhibit BMP among other signaling pathways, Noggin and LDN may have other types of activities which are different than inhibition of BMP.
  • a BMP inhibitor might be able to substitute for Noggin in differentiating cells of neural cell fate. Therefore, a small molecule BMP inhibitor, LDN-193189, (Yu, et al., Nat Med 14, 1363-1369, (2008), herein incorporated by reference) was used and found during the development of the present inventions to replace Noggin, in combination with SB431542, for generating primitive neuroectoderm from hPSCs, cells that have neural cell fate, i.e. CNS cells ( FIG. 2A ). This combination treatment was termed LSB for the combination of these two inhibitors LDN-193189 and SB431542.
  • cell differentiation was initiated by treatment of high confluency monolayer hES or hiPS with dual inhibition of SMAD signaling.
  • a preferred embodiment utilizes a percentage confluency of 50%400%, with a most preferred embodiment of 70%-80% confluency. It will be obvious to one skilled in the art that the initial plating density required to achieve a preferred confluency of the present invention will be dependent on cell type, size, plating efficiency, survival, adhesion and other parameters which can be determined empirically without undue experimentation on the part of the skilled artisan.
  • Dual inhibition of SMAD can be achieved with a variety of compounds including Noggin, SB431542, LDN-193189, Dorsomorphin, or other molecules that block TGF ⁇ , BMP, and Activin/Nodal signaling.
  • a preferred embodiment utilizes the composition comprising SB431542 and LDN-193189 (collectively, LSB) at a concentration of 0.1 ⁇ M-250 ⁇ M, or more preferable 1-25 ⁇ M, or most preferable 10 ⁇ M of SB431542 and 10-5000 nM, or most preferably 100-500 nM of LDN-193189.
  • DA-like neurons were used in transplantation studies that resulted in concerns on the further use of these cells for therapeutic applications.
  • procedures described in Perrier et al., 2004 and Fasano et al., 2010, including MS5 neural induction resulted in rosette cell formation and were used to make Day 11 precursors, see FIGS. 2, 16 and 17 for examples, that were further used to derive DA-like neurons.
  • These neurons resulted from a low percentage of the precursor cells in the resulting Day 11 cell populations.
  • FIG. 16A There were very small numbers of surviving TH+ neuron at 4.5 moths after transplantation ( ⁇ 50 TH+ cells/animal) in grafts from rosette derived DA neuron precursors FIG. 16A .
  • GFP marked cells GFP was driven by a ubiquitous promomoter
  • hNA+ green
  • co-express TH red
  • Panels 16 D-E show that D-E, despite the very poor in vivo survival there was some (low and highly variable) improvement in a few behavioral assays such as amphetamine induced rotations (D), cylinder test and spontaneous rotations (E).
  • Feeder-free neural induction was carried out as previously described (Chambers et al., 2009, herein incorporated by reference) but further modified to yield floor plate cells (Fasano et al., 2010, herein incorporated by reference).
  • the following example describes using exemplary cells from Section I for screening small molecule candidate compounds and determining whether their use would result in directed differentiation of a cell population containing a high percentage of midbrain floor plate neurons by Day 11 after the initial contact with the Dual-SMAD inhibitors.
  • the results of this screen initially showed that a SHH activating molecule together with activation of FGF8 and Wnt led to the efficient derivation of FOXA2+/LMX1A+positive midbrain floor plate cells from hESC by day 11 of differentiation.
  • the inventors show results herein of exemplary experiments that defined which molecules were necessary and the optimal time of contacting in order to derive the desired FOXA2+/LMX1A+positive cell population at Day 11.
  • CHIR99021 (CHIR) Induced a High Yield of Midbrain DA Neuron Precursor Fate by Day 11 of Culture.
  • CHIR99021 a potent GSK3 ⁇ inhibitor known to strongly activate WNT signaling, induced LMX1A in FOXA2+ floor plate precursors ( FIG. 1 a ).
  • CHIR was more potent than recombinant Wnt3A or Wnt1 at inducing LMX1A expression.
  • the efficiency of LMX1A induction was dependent on the timing of CHIR exposure with a maximum effect at day 3-11 ( FIG. 5 ).
  • Induction of FOXA2/LMX1A co-expression required strong activation of SHH signaling using purmorphamine, a small molecule agonist, alone or in combination with recombinant SHH ( FIG. 7 ).
  • FIG. 1 d were performed using global temporal gene expression profiling.
  • FOXA1, FOXA2 and several other SHH downstream targets including PTCH1 were amongst the most differentially regulated transcripts in LSB/S/F8/CHIR versus LSB treatment sets ( FIG. 1 e ).
  • LSB cultures were enriched for dorsal forebrain precursor markers such as HES5, PAX6, LHX2, and EMX2.
  • FIG. 1 f Direct comparison of LSB/S/F8/CHIR versus LSB/S/F8 treatment ( FIG. 1 f ) confirmed selective enrichment for midbrain DA precursor markers in LSB/S/F8/CHIR group and suggested hypothalamic precursor identity in LSB/S/F8 treated cultures based on the differential expression of RAX1, SIX3, and SIX6 (see also POMC, OTP expression in FIG. 2 d below).
  • the full list of differentially expressed transcripts Tables 1, 2 and gene ontology analysis FIG. 8 b (DAVID; http://david.abcc.ncifcrf.gov) confirmed enrichment for canonical WNT signaling upon CHIR treatment.
  • FIG. 19A shows examples of FOXA2/Tuj1 double labeled cells following LSB/S/F8/CHIR treatment (upper panels) and FOXA2 co-labeling with TH, Mull and LMX1A (lower panels). Those marker combinations are diagnostic for early stage midbrain DA neuron precursors.
  • FIG. 19B shows gene expression data (for comparison to FIG. 2E ) for key dopamine neuron precursor markers.
  • Human ESC(H9, H1) and iPSC lines (2C6 and SeV6) were subjected to a modified Dual SMAD-inhibition (Chambers, et al. Nat. Biotechnol. 27:275-280 (2009), herein incorporated by reference) based floor plate induction (Fasano, et al., Cell Stem Cell 6:336-347 (2010), herein incorporated by reference) protocol. Exposure to SHH C25II, Purmorphamine, FGF8 and CHIR99021 were optimized for midbrain floor plate and yield of novel populations of DA neuron (see FIG. 1 d ).
  • DA neuron survival and maturation factors Perrier, et al. Proc Natl Acad Sci USA 101:12543-8 (2004), herein incorporated by reference
  • AA AA
  • BDNF BDNF
  • GDNF GDNF
  • TGF ⁇ 3 TGF ⁇ 3
  • dbcAMP dbcAMP
  • DA neurons were injected stereotactically in the striata of the animals (150 ⁇ 10 3 cells in mice, 250 ⁇ 10 3 cells in rats) and a total of 7.5 ⁇ 10 6 cells (distributed in 6 tracts; 3 on each side of brain) in monkeys.
  • Behavioral assays were performed at monthly intervals post-grafting, including amphetamine mediated rotational analysis as well as a test for focal akinesia (“stepping test”) and limb use (cylinder test). Rats and mice were sacrificed at 18-20 weeks and the primates at 1 month post grafting. Characterization of the grafts was performed via stereological analyses of cell number and graft volumes as well as a comprehensive phenotypic characterization via immunohistochemistry.
  • hESC lines H9 WA-09, XX, passages 27-55 from when 10/2009
  • H1 WA-01, XY, passages 30-40 from when 6/2010
  • iPS cell lines 2C6 Kim, et al. Cell Stem Cell 8:695-706 (2011), herein incorporated by reference
  • XY passages 20-30
  • SeV6 XY, passages 20-30; derived from MRC-5 embryonic fibroblasts using non-integrating 4 factor Sendai vector system (Ban, et al. Proc. Natl. Acad. Sci.
  • LDN-193189 100 nM (ranging in concentration from 0.5-50 ⁇ M, Stemgent, Cambridge, Mass.), SB431542 (10 ⁇ M (ranging in concentration from 0.5-50 ⁇ M, Tocris, Ellisville, Mich.), SHH C25II (100 ng/ml (ranging in concentration from 10-2000 ng/ml, R&D, Minneapolis, Minn.), Purmorphamine (2 ⁇ M (ranging in concentration from 10-500 ng/ml, Stemgent), FGF8 (100 ng/ml (ranging in concentration from 10-500 ng/ml, R&D) and CHIR99021 (CHIR; 3 ⁇ M (ranging in concentration from 0.1-10 ⁇ M, Stemgent).
  • SHH treatment refers to exposure, i.e. contact, of cells to a combination of SHH C25II 100 ng/ml+Purmorphamine (2 ⁇ M).
  • Cells were plated (35 ⁇ 40 ⁇ 10 3 cells/cm 2 ) and cultured for 11 days on matrigel or geltrex (used as purchased) (BD, Franklin Lakes, N.J.) in Knockout serum replacement medium (KSR) containing DMEM, 15% knockout serum replacement, 2 mM L-glutamine and 10- ⁇ M (ranging in concentration from 1-25 ⁇ M ⁇ -mercaptoethanol.
  • KSR Knockout serum replacement medium
  • KSR medium was gradually shifted to N2 medium starting on day 5 of differentiation, by mixing in ratios of 75% (KSR):25% (N2) on day 5-6, 50% (KSR):50% (N2) day 7-8 and 25% (KSR):75% (N2) on day 9-10, as described previously (Chambers, et al. Nat. Biotechnol. 27:275-280 (2009), herein incorporated by reference).
  • media was changed to Neurobasal medium/B27medium (1:50 dilution)/L-Glut (effective ranges 0.2-2 mM)) containing medium (NB/B27; Invitrogen) supplemented with CHIR (until day 13) and with BDNF (brain-derived neurotrophic factor, 20 ng/ml ranging from 5 to 100; R&D), ascorbic acid (AA; 0.2 mM (ranging in concentration from 0.01-1 mM), Sigma, St Louis, Mich.), GDNF (glial cell line-derived neurotrophic factor, 20 ng/ml (ranging in concentration from 1-200 ng/ml); R&D), TGF ⁇ 3 (transforming growth factor type ⁇ 3, 1 ng/ml (ranging in concentration from 0.1-25 ng/ml); R&D), dibutyryl cAMP (0.5 mM (ranging in concentration from 0.05-2 mM); Sigma), and DAPT (10 nM (ranging in concentration from 0.5-50 nM); Tocris,
  • cells were dissociated using Accutase® (Innovative Cell Technology, San Diego, Calif.) and replated under high cell density conditions (for example from 300-400 k cells/cm 2 ) on dishes pre-coated with polyornithine (PO); 15 ⁇ g/ml (ranging in concentration from 1-50 ⁇ g/ml)/Laminin (1 ⁇ g/ml) (ranging in concentration from 0.1-10 ⁇ g/ml)/Fibronectin (2 ⁇ g/ml (ranging in concentration from 0.1-20 ⁇ g/ml) in differentiation medium (NB/B27+BDNF, AA, GDNF, dbcAMP (ranging in concentration as described herein), TGF ⁇ 3 and DAPT (ranging in concentration as described herein) until the desired maturation stage for a given experiment.
  • Accutase® Innovative Cell Technology, San Diego, Calif.
  • hESCs were induced towards neural fate by coculture with irradiated MS5 cells in KSR supplemented with SB431542 and Noggin (250 ng/ml (ranging in concentration from 10-1000 ng/ml); R&D), from day 2-8 and SHH+FGF8 from day 6-11 of differentiation.
  • neural rosettes were manually isolated and cultured (P1 stage) in N2 medium supplemented with SHH, FGF8, BDNF and AA as described previously (Perrier, et al. Proc Natl Acad Sci USA 101:12543-8 (2004), herein incorporated by reference). After 5-7 days in P1 stage, rosettes were again harvested mechanically and triturated following incubation in Ca 2 /Mg 2 -free Hanks' balanced salt solution (HBSS) for 1 h and replated on polyornithine (P0)/Laminin/Fibronectin coated plates.
  • HBSS Hanks' balanced salt solution
  • Patterning with SHH/FGF8 was continued for 7 days at P2 stage followed by final differentiation in the presence of BDNF, AA, GDNF, TGFb3 and dbcAMP as described above until the desired maturation stage for a given experiment (typically 5-7 days for transplantation studies or 32 days for in vitro functional studies).
  • RNeasy kit Qiagen, Valencia, Calif.
  • RNA at day 25 of each condition was reverse transcribed (Quantitech, Qiagen) and amplified material was detected using commercially available Taqman gene expression assays (Applied Biosystems, Carlsbad, Calif.) with the data normalized to HPRT. Each data point represents 9 technical replicates from 3 independent biological samples.
  • Raw data of microarray studies are not yet available at GEO worldwideweb.ncbi.nlm.nih.gov/geo).
  • Rodent and monkey procedures were performed following NIH guidelines, and were approved by the local Institutional Animal Care and Use Committee (IACUC), the Institutional Biosafety Committee (IBC) as well as the Embryonic Stem Cell Research Committee (ESCRO).
  • IACUC Institutional Animal Care and Use Committee
  • IBC Institutional Biosafety Committee
  • ESCRO Embryonic Stem Cell Research Committee
  • NOD-SCID IL2Rgc null mice (20-35 g in weight; Jackson Laboratory, Bar Harbor, Me.) were anesthetized with Ketamine (90 mg/kg; Akorn, Decatur, Ill.) and Xylazine (4 mg/kg Fort Dodge, Iowa). 6-hydroxydopamine (ranging in concentration from 1-20 ⁇ g) 6-OHDA (Sigma-Aldrich) was injected stereotactically into the striatum at the following coordinates (in millimeters): AP, 0.5 (from bregma; a skull suture used as reference for stereotactic surgery); ML, ⁇ 2.0; DV, ⁇ 3.0 (from dura a membrane covering the brain used for reference).
  • mice with successful lesions were selected for transplantation.
  • a total of 150 ⁇ 10 3 cells were injected in a volume of 1.5 ⁇ l into the striatum at the following coordinates (in mm): AP, 0.5; ML, ⁇ 1.8; DV, 3.2.
  • the mice were sacrificed 18 weeks post transplantation.
  • Rats were anesthetized with Ketamine (90 mg/kg) and xylazine (4 mg/kg) during surgical procedures. Unilateral, medial forebrain bundle lesions of the nigro-striatal pathway were established by stereotaxic injection of 6-OHDA (3.6 mg/ml in 0.2% ascorbic acid and 0.9% saline, Sigma) at two sites (Studer, et al. Nature Neurosci. 1:290-295 (1998), herein incorporated by reference). Rats were selected for transplantation if amphetamine-induced rotation exceeded 6 rotations/min by 6-8 weeks post injection.
  • Bilateral injections of cells (10 ul/injection; 125,000 cell/ul were performed at three sites (1-posterior caudate, 2-pre-commissural putamen and overlying white matter) for a total volume of 30 ⁇ l per hemisphere.
  • An infusion pump attached to a stereotaxic micromanipulator was utilized to deliver the cells at a rate of 1 ⁇ l/min though a 50 ⁇ l Hamilton syringe with 28 G needle. After the injections were completed, the needle was left in place for an additional 2-5 minutes to allow the infusate to diffuse off the needle tip before slowly retracting the syringe.
  • the animals received analgesics (buprenex, 0.01 mg/kg IM, BID for 72 hours post surgery; meloxicam, 0.1 mg/kg SQ, SID for 72 hours post surgery) as well as an antibiotic (cephazolin, 25 mg/kg IM, BID) until 72-hours post-surgery.
  • analgesics buprenex, 0.01 mg/kg IM, BID for 72 hours post surgery; meloxicam, 0.1 mg/kg SQ, SID for 72 hours post surgery
  • antibiotic cephazolin, 25 mg/kg IM, BID
  • the animals received cyclosporine A (Neoral, Sandimmune) orally (30 mg/kg tapered to 15 mg/kg) once daily beginning 48-hrs prior to surgery until sacrifice, one month following transplantation.
  • Amphetamine-induced rotations (mice and rats) and the stepping test (rat) were carried out before transplantation and 4, 8, 12, 18 weeks after transplantation.
  • Rotation behavior in mice was recorded 10 min after i.p. injection of d-amphetamine (10 mg/kg, Sigma) and recorded for 30 minutes.
  • Rotation behavior in rats was recorded 40 min after i.p. injection of d-amphetamine (5 mg/kg) and automatically assessed by the TSE VideoMot2 system (Germany). The data were presented as the average number of rotations per minute.
  • the stepping test was modified from Blume, et al. Exp. Neurol. 219:208-211 (2009) and Crawley, et al.
  • the cylinder test was performed by placing each animal in a glass cylinder and counting the number of ipsilateral versus contralateral paw touches (out of 20 touches) to the wall of the cylinder as described previously (Tabar, et al. Nature Med. 14:379-381 (2008), herein incorporated by reference). Tissue Processing for rodents and primates are described below.
  • ketamine (10 mg/kg, Intramuscular (IM)) and pentobarbital (25 mg/kg, intravenous (IV)) via cardiac perfusion with heparinized 0.9% saline followed by fresh cold 4% PFA fixative (pH7.4).
  • brains were removed from the skull and post-fixed in 4% PFA, free-floating, for 24-36 hrs. They were then rinsed and re-suspended in 10% sucrose on a slow shaker at 4° C., and allowed to “sink”. The process was then repeated in 20% sucrose followed by 30% sucrose.
  • Whole brains were cut coronally into 40 ⁇ m serial sections on a frozen sledge microtome and stored free-floating in cryopreservative medium at ⁇ 20° Celcius.
  • HVA Homovanillic acid
  • DOPAC 3,4-Dihydroxy-Phenylacetic Acid
  • Cultures were transferred to a recording chamber on an upright microscope equipped with a 40 ⁇ water-immersion objective (Eclipse E600FN; Nikon); cultures were perfused with saline containing in mM: 125 NaCl, 2.5 KCl, 25 NaHCO 3 , 1.25 NaH 2 PO 4 , 2 CaCl, 1 MgCl 2 , and 25 glucose (34° C.; saturated with 95% 0 2 -5% CO 2 ; pH 7.4; 298 mOsm/L).
  • the saline flow rate was 2-3 ml/min running through an in-line heater (SH-27B with TC-324B controller; Warner Instruments).
  • Neurons were visualized by video microscopy with a cooled-CCD digital camera (CoolSNAP ES 2 , Photometrics, Roper Scientific, Arlington, Ariz.). Cells selected for electrophysiological recordings had neuron-like shapes with fine branching neurites. Somatic whole-cell patch-clamp recordings in current clamp configuration were performed with a MultiClamp 700B amplifier (Molecular Devices). Signals were filtered at 1-4 kHz and digitized at 5-20 kHz with a Digidata 1440A (Molecular Devices). Recording patch electrodes were fabricated from filamented borosilicate glass (Sutter Instruments) pulled on a Flaming-Brown puller (P-97, Sutter Instruments) and had resistances of 4-6 M ⁇ in the bath.
  • Electrodes were filled with internal solution containing in mM: 135 K-MeSO 4 , 5 KCl, 5 HEPES, 0.25 EGTA, 10 phosphocroeatine-di(tris), 2 ATP-Mg, and 0.5 GTP-Na (pH 7.3, osmolarity adjusted to 290-300 mOsm/L).
  • the amplifier bridge circuit was adjusted to compensate for electrode resistance and monitored. Electrode capacitance was compensated. When series resistance increased >20% during the recording, the data were discarded because increased resistance suggested a partial technical failure during recordings.
  • FIGS. 3 and 11 The percentages of marker positive cells at the floor plate (day 11) FIG. 1 , midbrain dopamine neuron precursor (day 25), FIG. 2 and mature DA neuron stages (day 50 or later) FIGS. 3 and 11 , were determined in samples derived from at least 3 independent experiments each. Images for quantification were selected in a uniform random manner and each image was scored first for the number of DAPI-positive nuclei, followed by counting the number of cells expressing the marker of interest. Data are presented as mean ⁇ SEM. Quantification of human cells (identified with anti-hNA) and TH+ neurons within grafts was performed on every tenth section where a graft was identifiable.
  • hESC medium for maintenance (1 liter): 800 mL DMEM/F12, 200 mL of Knockout Serum Replacement, 5 mL of 200 mM L-Glutamine, 5 mL of Pen/Strep, 10 mL of 10 mM MEM minimum non-essential amino 15 acids solution, 55 ⁇ M of 13-mercaptoethanol, and bFGF (final concentration is 4 ng/mL).
  • KSR medium for hESC differentiation (1 liter): 820 mL of Knock out DMEM, 150 mL of Knock out Serum Replacement, 10 mL of 200 mM L-Glutamine, 10 mL of Pen/Strep, 10 mL of 10 mM MEM, and 55 ⁇ M of 13-mercaptoethanol.
  • DMEM Dulbecco's Modification of Eagles Medium
  • FBS primary mouse embryo fibroblast
  • FBS primary mouse embryo fibroblast
  • Day 13 is a midbrain floor plate stage, characterized by co-expression of FOXA2/LMX1A. In addition to expression of FOXA2 and LMX1A, a loss of OCT4 expression and lack of induction of forebrain markers PAX6 and FOXG1 is found.
  • Day 25 is the midbrain DA neuron precursor stage, characterized by the continuous expression of FOXA2/LMX1A and expression of the neuronal (TUJ1) and DA markers (NURR1, TH).
  • Proliferating, Ki67+ cells and the number of PAX6 and FOXG1 forebrain neural precursors are monitored, where these markers are not desired.
  • an Operetta (Perkin Elmer) High Content Microscope was used for measurements.
  • qRT-PCR assays was also used for each marker to confirm immunofluorescence data.
  • cell lines (cultures) passing these preliminary in vitro tests are used for engraftment, see, Table 7.
  • mature DA neurons were cryopreserved without serum at day 25 (ranging from day 20-day 25) in culture medium +7% DMSO (ranging from 3%-12%) until thawed for use in engraftment.
  • cell samples are stored in liquid nitrogen.
  • cells are stored in low temperature freezers.
  • cryoprotectants such as myoinositol, polyvinyl alcohol, serum replacement, caspase inhibition compounds, are contemplated for use in addition to DMSO. After thawing, cells are tested for viability, marker expression, etc., prior for use in grafting. In some embodiments, thawed cells were tested for maintenance of function in long-term in vitro and in vivo assays for monitoring freezing and storage conditions.
  • tissue culture components are contemplated for use in producing cells of the present inventions.
  • FGF8 was shown that although its use resulted in cells of the present inventions, it was not required for production of these cells.
  • additional reagents such as those listed in Table 8, as additives to cell cultures along with the four “core” molecules that resulted in DA neuronal cells of the present inventions, i.e.
  • SB431542 and LDN193189 showed efficient neural conversion of pluripotent stem cells while the addition of Purmorphamine and CHIR99021 to these cells demonstrated midbrain floor plate induction.
  • Other chemicals and recombinant proteins were or can be used to provide long-term trophic support and/or accelerate differentiation. Some of these compounds will be used in further tests in order to define their roles in cell differentiation described herein.
  • a cell number threshold is contemplated for determining a clinically relevant contamination limit of problematic cell types, such as undifferentiated hESCs which may develop into teratomas or primitive neuroectodermal precursors capable of significant proliferation. Therefore, some embodiments are contemplated for further enrichment of dopaminergic neurons in cells for use in grafts, i.e. depleting contaminating cell types prior to engraftment, see, Table 7 for exemplary limits.
  • hESCs For hESCs, a pre-determined mix of undifferentiated (Oct4+/Nanog+) cells with hES derived DA neurons will be used to monitor clinical symptoms suggestive of mass effect and/or animal death in animal experiments. A dose response of one hES cell per 10,000 hESC-derived DA cells, 1/5000, 1/1000 and 1/100 will be performed. Cells will be injected intra-striatally and the animals will receive immunosuppression. Rats will be monitored closely and will be sacrificed upon manifestation of neurological symptoms or at a maximum of 6 months. The brains will be analyzed for graft volumes and composition as described herein.
  • the cell ratios will adjusted until a clear in vivo threshold is established for the emergence of teratomas. For determining contaminating levels of primitive neuroectodermal precursors, a similar strategy will be followed.
  • the presence of early neural precursors have a significant potential for proliferation and broad differentiation into central nervous system as well as peripheral nervous system (PNS) fates.
  • PNS peripheral nervous system
  • Graft analysis will consist of IHC for rosette cells (PLZF expression), their CNS progeny (neural precursors expressing Nestin/Sox2 or forebrain precursors, expressing FoxG1) as well as graft volumes and a proliferation index (% Ki67+ of total surviving cells).
  • Parkinson's disease is the second most common neurodegenerative disorder and is estimated to affect 4.1-4.6 million patients world-wide, a number predicted to more than double by 2030. It is the second most common neurodegenerative disorder after Alzheimer's disease, affecting approximately 1 million patients in the US with 60,000 new patients diagnosed each year. The disease has a major socioeconomic impact causing significant morbidity and mortality, and the combined direct and indirect costs of PD, including health care cost and lost income, is estimated to be approximately $25 billion per year in the US alone. Currently there is no cure for Parkinson's disease (PD), an age-related, progressive and disabling disorder. PD is characterized pathologically by a selective loss of midbrain DA neurons in the substantia nigra.
  • a fundamental characteristic of PD is therefore progressive, severe and irreversible loss of midbrain dopamine (DA) neurons resulting in ultimately disabling motor dysfunction.
  • DA midbrain dopamine
  • pharmacological, exercise-based, gene- and surgical therapies have been developed for PD, none of those approaches are yet able to restore proper DA neuron function.
  • Long-term control of motor symptoms in patients often remains suboptimal, and while recognizing the importance progressive non-dopamine responsive motor and non-motor symptoms, the fundamental issue of long term dopamine-responsive symptom control remains an area of critical therapeutic need.
  • PD widespread pathology
  • the cardinal features of PD (bradykinesia, rigidity, and tremor partially) are fundamentally related to DA neuronal cell loss and are dopamine-responsive.
  • PD is contemplated for treatment using neuronal cell replacement due to the rather selective loss of midbrain DA neurons that is responsible for most motor symptoms of the disease.
  • a healthy human brain harbors approximately one million DA neurons. Therefore, in one embodiment, DA neuron replacement is contemplated to require a relatively small number of surviving cells as compared to most other disorders in the CNS.
  • fetal tissue transplantation fails to replace DA neuronal function.
  • fetal tissue transplantation is plagued by multiple challenges including low quantity and quality of donor tissue, ethical and practical issues surrounding tissue acquisition, and the poorly defined heterogeneous nature of transplanted cells, which are some of the factors contributing to the variable clinical outcomes. Examples of fetal transplantation are described in; Mendez, et al. Nature Med. (2008)); Kordower, et al. N. Engl. J. Med. 332:1118-1124 (1995); Piccini, et al. Nature Neuroscience 2:1137-1140 (1999).
  • a stem cell-derived cell source or other type of consistent cell type for use in providing cells for transplantation is contemplated to overcome many of the challenges associated with fetal tissue grafting and could offer an unlimited source of DA neurons at the optimum stage for transplantation.
  • the inventors' succeeded in obtaining authentic human midbrain DA neurons from pluripotent stem cells capable of reversing neurological defects in murine animals and primates.
  • This novel differentiation strategy was highly efficient and led to robust in vivo engraftment of the cells, induction of functional recovery in PD models of disease, and lack of adverse events such as inappropriate cell proliferation as supported by preclinical data.
  • Additional embodiments including “enhancement” strategies to control cell purity promote axonal fiber outgrowth and include novel safety/regulatory features in grafting strategies are contemplated.
  • a method of cell purification demonstrated starting from a simple surface marker screen against a cell type of interest, (such as CD142) towards a meaningful enrichment strategy for a specific neuron type.
  • this method is contemplated for use in providing cells for use in humans.
  • engineered expression of PSA-NCAM is contemplated for enhancing axonal outgrowth for use in neural repair in vivo.
  • Such applications include promotion of long-distance axonal growth for treating motoneuron disease, Huntington's disease or other disorders primarily affecting projection neurons. Additional embodiments are contemplated for a GMP qualified pluripotent cell source, and the like. Because the engraftment methods described herein, require a small number of DA neurons and are based upon relatively simple, cost-effective small molecule methods developed for DA neuron induction, it is contemplated that DA neuron replacement therapy would be at a reasonable cost on a per patient basis.
  • PD neuronal degeneration in PD proceeds to affect many cell types other than midbrain dopamine neurons, particularly at later stages of the disease.
  • non-DA responsive symptoms predominate in late PD, leading to dysphagia, falls, dementia and other significant morbidities.
  • some non-motor symptoms are contemplated to benefit from restoring dopaminergic function.
  • the use of hESC derived DA neurons at early stages of the disease would prevent some of the secondary PD symptoms, including the degeneration of the dopamine receptive populations of the striatum.
  • One major benefit of using mature DA neuron cell engraftment therapy of the present inventions is the unique neurorestorative nature post engraftment, i.e. long term recovery of neuronal function that is contemplated for use in patients for progressive removal of drug therapy.
  • Cell transplantation is contemplated to affect a different spectrum of DA-related symptoms than those responding to drugs or other therapy.
  • mature DA neuron transplantation is contemplated for use with DBS.
  • mature DA neuron is contemplated for use with therapy.
  • Pulsatile delivery of L-dopa has a major role in development of these later stage motor complications therefore a “smoother” more physiologic delivery of dopamine, i.e. such as from engrafted cells of the present inventions, would therefore be highly desirable.
  • pharmacological strategies there are several surgical treatment options. These include the ablation or functional inactivation of cells within the basal ganglia via pallidotomy or deep brain stimulation by targeting the subthalamic nucleus or globus pallidus pars interna. While these surgical options are alternatives for some patients, they provide symptomatic relief from the disease but do not restore normal DA function.
  • DA neuron replacement in PD was done in the 1980s based on the use of adrenal medulla derived chromaffin cells. Those hormone producing cells were shown to switch neurotransmitter phenotype from adrenalin to DA when placed ectopically into the CNS.
  • GID graft-induced dyskinesias
  • fetal grafting treatment Another problem with fetal grafting treatment was (and is) limited availability of fetal midbrain tissue at the appropriate developmental stage.
  • An alternative strategy was tried clinically to address the issue of limited supply by using fetal pig derived DA neurons.
  • DA neuron survival in those xenografts was poor and the overall approach was abandoned.
  • a recent trial using retinal pigmented epithelium also failed to show any benefits.
  • the use of human ES cells as sources of transplant cells is contemplated to provide an unlimited source of cells for making dopamine neurons for use in transplantation.
  • timing of CHIR99021 exposure determines induction of FOXA2/LMX1A midbrain floor plate precursors. Therefore the inventors tested for immunocytochemical analysis of FOXA2/LMX1A at day 11 of differentiation following LSB/S/F8 (i.e. treating cells with LSB, S, i.e. SHH, and FGF8 (F8) treatment alone or in combination with CHIR starting at various timepoints: d(day)0-d11, d1-d11, d3-d11, d5-d11, d7-d11 compared to duplicate cultures of cells with no CHIR treatment. Then quantification of the percentage of FOXA2+, LMX1A+ and double labeled cells were determined at day 11 of differentiation following differential onset of CHIR exposure as described in the immunocytochemical analysis.
  • CHIR99021 (C) is a Factor for Inducing FOXA2+/LMX1A+ Cells by Day 11 from LSB Cultured Cells Contacted with an Activator of Hedgehog and Purmorphamine.
  • the following example describes using exemplary methods for testing the efficacy of each compound for inducing directed neuronal differentiation of mDA neurons.
  • a cell population containing pluripotent cells was chosen by the inventors for a starting population and plated at Day 0.
  • the inventors followed a cell population with regular feedings containing fresh LSB until Day 11 and discovered that some remaining cells were LMX1A+ but did not express FOXA2 ( FIG. 1 a,b ).
  • the inventors plated duplicate starting cell populations then tested for cell types (i.e.
  • Three primary exemplary culture conditions tested were 1) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11, 2) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11 while during this time cells were additionally contacted with fresh purmorphamine, SHH and FGF8 until Day 7 and 3) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11 while during this time cells were additionally contacted with fresh purmorphamine, SHH and FGF8 until Day 7 while additionally contacted with fresh CHIR starting on Day 3 of culture until Day 11 with several variations of these primary conditions in order to determine optimal yield of cell types.
  • FIG. 1 d Systematic comparisons of the three culture conditions ( FIG. 1 d ) were performed using global temporal gene expression profiling. Hierarchical clustering of differentially expressed genes segregated the three treatment conditions by day 11 of differentiation ( FIG. 8 a ).
  • FOXA1, FOXA2 and several other SHH downstream targets including PTCH1 were amongst the most differentially regulated transcripts in LSB/S/F8/CHIR versus LSB treatment sets ( FIG. 1 e ).
  • LSB cultures were enriched for dorsal forebrain precursor markers such as HES5, PAX6, LHX2, and EMX2.
  • Direct comparison of LSB/S/F8/CHIR versus LSB/S/F8 treatment confirmed selective enrichment for midbrain DA precursor markers in LSB/S/F8/CHIR group and suggested hypothalamic precursor identity in LSB/S/F8 treated cultures based on the differential expression of RAX1, SIX3, and SIX6 (see also POMC, OTP expression in FIG. 2 d below).
  • An exemplary list of differentially expressed transcripts are shown, i.e. Tables 1, 2 and gene ontology analysis for Day 11, FIG.
  • LSB dorsal forebrain
  • LSB/S/F8 ventral/hypothalamic
  • LSB/S/F8/CHIR midbrain DA precursor identity
  • precursor FOXA2+/LMX1A+ cells were maintained in a medium promoting neuronal maturation (BAGCT, see Example I).
  • BAGCT neuronal maturation
  • two other techniques were used to generate DA neuronal precursor cells.
  • the following types of comparisons were made between the populations of differentiated cells resulting from previous methods and methods of the present inventions: A) Immunocytochemical analysis at day 50 of differentiation for TH in combination with LMX1A, FOXA2 and NURR1, B) Quantification of TH+, FOXA2+, LMX1+, and NURR1+ cells out of total cells comparing rosette-derived versus floor plate-derived (LSB/S/F8/CHIR) cultures.
  • FIG. 2 a Three precursor cell populations yielded Tuj1+ neurons ( FIG. 2 a ) and cells expressing TH, the rate-limiting enzyme in the synthesis of DA.
  • LSB/S/F8/CHIR treatment yielded TH+ cells that co-expressed LMX1A and FOXA2 and displayed strong induction of the nuclear receptor NURR1 (NR4A2) ( FIG. 2 a,b ).
  • Comparing gene expression in day 13 versus day 25 cultures confirmed robust induction of other postmitotic DA neuron markers ( FIG. 2 c ). Characterizing DA neuron identity at day 25 in comparison to LSB and LSB/S/F8 treated cultures confirmed enrichment for known midbrain DA neuron transcripts and identified multiple novel candidate markers ( FIG.
  • the transcript most highly enriched in LSB/S/F8/CHIR was TTF3, a gene not previously associated with midbrain DA neuron development, but highly expressed in the human substantia nigra ( FIG. 8 c ; Allen Brain Atlas: http://human.brain-map.org).
  • FIGS. 10 and 16 In vitro and in vivo properties of floor plate-derived DA neurons were compared to DA-like neurons obtained via a neural rosette intermediate ( FIGS. 10 and 16 ). Patterning of neural rosettes represents the currently most widely used strategy for deriving DA neurons from hPSCs. Both floor plate- and rosette-based protocols were efficient at generating TH+ neurons capable of long-term in vitro survival (day 50 of differentiation; FIG. 3-1 a ). However, the percentage of TH+ cells was significantly higher in floor plate-derived cultures ( FIG. 3-1 b ). While TH+ cells in both protocols displayed co-expression of NURR1, floor plate-derived DA neurons co-expressed FOXA2 and LMX1A ( FIG. 3-1 a,b and 3-2).
  • FIG. 3-1 c Few GABA and serotonin (5-HT)-positive neurons were observed ( FIG. 3-1 c ).
  • DA and its metabolites DOPAC and HVA, were present in cultures generated with either protocol, but DA levels were approximately 8 times higher in floor plate cultures ( FIG. 3-1 d,e ).
  • Midbrain DA neurons exhibited extensive fiber outgrowth and robust expression of mature neuronal markers including synapsin, dopamine transporter (DAT), and G-protein coupled, inwardly rectifying potassium channel (Kir3.2, also called GIRK2, expressed in substantia nigra pars compacta (SNpc) DA neurons) ( FIG. 3-1 f , FIG. 11 ).
  • SNpc DA neurons in vivo exhibit an electrophysiological phenotype that differentiates them from most other neurons in the brain. In particular, they spike spontaneously at a slow (1-3 Hz) rate. Moreover, this slow spiking is accompanied by a slow, sub-threshold oscillatory potential. After 2-3 weeks in vitro, these same physiological features are displayed by SNpc DA neurons cultured from early postnatal mice. The DA neurons differentiated from hESCs consistently (4/4 tests) displayed this distinctive physiological phenotype ( FIG. 3-1 g - i ).
  • FIG. 3-1 j DA release measurement by HPLC showed d65 old TH+ neurons are functional in vitro FIG. 3-1 k.
  • Floor plate derived DA neurons were obtained from human ES cells following small molecule based activation of SHH and canonical WNT signaling during early differentiation stages ( FIG. 3-2 ). These hES cells progressed from a FOXA2/LMX1A double positive midbrain floor plate stage, to Tuj1+ immature neurons with co-expression of FOXA2/LMX1A then to mature DA neurons ( FIG.
  • the following example describes using exemplary methods of the present inventions for use in therapeutic cell replacement.
  • One major challenge in the field is the ability to generate hPSC-derived midbrain DA neurons that functionally engraft in vivo without the risk of neural overgrowth or inappropriate differentiation into non-midbrain neurons or develop teratomas.
  • the time of cell cycle exit marked by expression of NURR1
  • the time of cell cycle exit may be a suitable stage for grafting (approximately day 25 of differentiation, FIG. 2 ).
  • Initial studies using day 25 cells in non-lesioned adult mice showed robust survival of hPSC-derived FOXA2+/TH+ neurons at 6 weeks after transplantation ( FIG. 12 ).
  • the overgrowth was likely due to the longer survival periods (4.5 months versus 6 weeks), lack of FACS purification prior to transplantation and choice of NOD-SCID IL2Rgc null host.
  • the number of proliferating Ki67+ cells was minimal in floor plate-derived grafts ( ⁇ 1% of total cells), while rosette-derived grafts retained pockets of proliferating neural precursors.
  • Neural overgrowth is thought to be caused by primitive anterior neuroectodermal cells within the graft (Elkabetz, et al. Genes Dev. 22:152-165 (2008); Aubry, et al. Proc. Natl. Acad. Sci. U.S.A. 105:16707-16712 (2008), herein incorporated by reference).
  • results in NOD-SCID IL2Rgc null mice described herein demonstrated robust long-term survival of FOXA2+/TH+ neurons, complete reversal of amphetamine-induced rotation behavior and lack of any signs of neural overgrowth. However, some of these outcomes could be attributable to the specific use of NOD-SCID IL2Rgc null mice.
  • floor plate-derived DA neuron cultures 250 ⁇ 10 3 cells were transplanted in adult 6-OHDA lesioned rats immunosuppressed pharmacologically using cyclosporine A. Five months after transplantation graft survival was robust ( FIG. 4 e - h ) with an average of more than 15,000 TH+ cells co-expressing FOXA2 ( FIG.
  • FIG. 4 g TH+/hNCAM+ fibers emanated from the graft core into the surrounding host striatum ( FIG. 4 f ).
  • TH+ cells expressed midbrain DA neuron markers PITX3 and NURR1 ( FIG. 4 h - j ).
  • Behavioral analyses showed complete rescue of amphetamine-induced rotational asymmetry, in contrast to sham-grafted animals that did not show improvements ( FIG. 4 k ). Grafted animals also showed improvements in the stepping test ( FIG. 4 l ) measuring forelimb akinesia and in the cylinder test ( FIG.
  • mice As in mice ( FIG. 13 ), serotonergic and GABAergic cells were rare ( ⁇ 1% of total cells) in rat cells, as were the mostly host-derived GFAP+ glial cells (7% of total cells; ( FIG. 14 ). While few serotonin+ neurons were detected in the graft, hNCAM-negative cells were observed that were likely host-derived serotonergic fibers ( FIG. 14 ).
  • Engraftment of floor-plate derived DA neurons in mice, rats, and monkeys demonstrated the surprising recovery of neuronal function in rodent and primate species.
  • Short-term (6 weeks) survival assays were extended for surprisingly long-term survival for up to 5 months after transplantation into the mouse striatum of 6OHDA lesioned and immunocompromised host mice.
  • a direct comparison of a traditional, rosette-based method of making DA neurons (Perrier, et al. Proc Natl Acad Sci USA 101, 12543-8 (2004)) compared to the novel floor plate based DA neuron differentiation protocol described herein, showed that floor plate derived DA neurons were capable of long-term DA neuron engraftment while rosette-based neurons were not ( FIG. 4 a - c ).
  • batches of 50 ⁇ 10 6 transplantable DA neuron precursors were obtained by day 25 of differentiation using the floor plate-based protocol.
  • Classic dose was 3 mg MPTP-HCL injected into the carotid artery (range 0.5-5 mg). This was followed by systemic injection of MPTP 0.2 mg/kg IV of MPTP.
  • Cells were injected at three locations (posterior caudate and pre-commissural putamen) on each side of the brain (6 tracts in total, 1.25 ⁇ 10 6 cells/tract), and the animals were immunosuppressed with cyclosporine-A.
  • the graft cores were composed of TH+ neurons co-expressing SC-121 ( FIG. 4 s ) and FOXA2 ( FIG. 4 t ).
  • SC-121 and GFP negative areas within the graft contained Iba1+ host microglia ( FIG. 15 ) indicating incomplete immunosuppression.
  • MPTP MPTP-HCL (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; ranging in concentration from 0.5-5 mg MPTP-HCl) lesioned rhesus monkeys containing a severe >95% loss of endogenous midbrain DA neurons.
  • MPTP exposure caused observable changes and symptoms similar to Parkinson's disease in humans.
  • duration of survival and extent of behavioral assessments For short term studies e.g. aiming at confirming cell survival or phenotypic composition, behavioral assessments are contemplated and the animals will be tested about 4-8 weeks post grafting for survival. Long-term studies will include behavioral assessment post grafting and animal survival for at least 5 months following grafting.
  • behavioral assessment are contemplated to include more complex parameters such as the staircase test for skilled forelimb use.
  • Exemplary protocols for assessment of in vivo performance has at least four main components and includes the following: i—Lesion Induction, ii—Core Behavioral Analysis, iii Grafting, and iv—Tissue Analysis. Although these procedures were used on rats, in some embodiments these procedures may be use on other species.
  • 6-hydroxydopamine 6-hydroxydopamine
  • MFB median forebrain bundle
  • 6-OHDA 6-hydroxydopamine
  • Targeted neurons include the A9 dopaminergic neurons within the substantia nigra compacta (SNC) as well as the A10 neurons in the ventral tegmental area.
  • This lesion model is widely studied and accepted as an excellent pre-clinical model for the study of the neurochemical and behavioral consequences of advanced PD as it results in extensive unilateral depletion of dopamine within the caudate-putamen complex (CPU).
  • the behavioral consequences are also well described and include spontaneous and drug-induced rotations as well as impairment in limb use (see below).
  • Bilateral models of Parkinsonism may better mimic human disease but they result in adipsia and aphagia in rats.
  • the procedure was performed in anesthetized animals (Ketamine/xylazine) via stereotactic injection in 2 sites along the median forebrain bundle.
  • the efficiency of complete lesion induction is highly dependent on the experience of the operator and during development of the present inventions ranged from 60-80%.
  • the animals were allowed to recover and then subjected to a battery of behavioral tests starting 2 weeks after surgery.
  • Behavioral analysis is initiated 2 weeks following surgery and continues after transplantation until the animal is sacrificed. Its purpose is 1) to establish that the lesion is stable and complete and that the animal has not exhibited spontaneous reversal of the symptoms, a phenomenon that has been shown in partially lesioned animals, 2) to demonstrate the impact of transplantation of dopamine neurons on established behavioral parameters.
  • Rats are observed for spontaneous rotations and for D-amphetamine-induced (ipsilateral) rotations (10 mg/kg). A threshold of >6 rotations per min is required as an indicator for a significant lesion. Apomorphine-induced rotations can also be analyzed but positive results are contemplated to require >80-90% depletion of dopamine innervation in the caudate-putamen and are less consistent in MFB lesions, in comparison to CPU lesions. Three sets of data are obtained at 2 week intervals and averaged. Rats with a rotation score of ⁇ 6 are not included in the studies.
  • Animals receive stereotactic injections of dopamine neurons into the striatum, at 3 weeks following the Parkinsonism-inducing lesions, and if behavioral testing confirms an adequate lesion.
  • the coordinates for injection are widely established.
  • the same set of behavioral tests is performed bimonthly for variable durations (on average 5 months are required to achieve stable behavioral recovery).
  • Antibodies include TH, FoxA2, Pitx3, Nurr1, Lmx1a, Girk2, DAT for dopamine neuron identity and function; 5-HT to identify Serotonergic neurons; Human NCAM or human nuclear antigen for human identity; Nestin, Sox2 for neural precursors; Ki-67 for proliferation; Oct4, Nanog for pluripotency markers; alpha-fetoprotein; myosin; cytokeratin for multi-lineage markers to rule out teratomas formation.
  • Quantitative parameters include graft volumes (Cavalieri estimator), total cell counts and dopaminergic cell counts (using TH/FoxA2 double labeling neurons) and Proliferative index (% Ki67+). Antibodies listed are commercially available and used during the development of the present inventions.
  • SD rats are contemplated for use in order to better model the human situation.
  • SD rats will receive daily intraperitoneal injections of cyclosporine (15 mg/kg) starting one day prior to grafting until sacrifice.
  • cyclosporine 15 mg/kg
  • exemplary methods for scaling up mDA neuron cultures are provided, see, Table 9. In particular embodiments, such methods are contemplated for use in producing GMP level cultures for clinical use.
  • cell sources for use in differentiation methods of the present inventions include but are not limited to WA09, ACT (M09), Bio-Time and Roslin cell lines. In some embodiments, cell sources for use in differentiation methods of the present inventions include but are not limited to GMP grade lines. In some embodiments, cell sources for use in differentiation methods of the present inventions include but are not limited to production of mature DA neurons for use in short term engraftment survival analysis. In some embodiments, cell sources for use in differentiation methods of the present inventions include but are not limited to production of mature DA neurons for use in engraftment experiments that include behavioral assessments. Controls will consist of the Research Grade WA09 and a sham saline group. Statistical analyses will use ANOVA with the Dunnett post-hoc test.
  • Standard behavioral tests (as described herein) exhibited a direct correlation with the number of surviving dopamine neurons within the graft.
  • behavior tests show maximum recovery with an estimate of about 30% DA neuron recovery which is required to reduce amphetamine-induced rotations.
  • a threshold for survival of DA neurons in the host is reached (an estimate of 800-1200 DA neurons in the rat model)
  • a microtransplantation approach was described that results in placement of multiple small grafts throughout the striatum, as opposed to large grafts.
  • mature DA neurons of the present inventions are administered to one location of the striatum.
  • mature DA neurons of the present inventions are administered to at least 2 or more locations within the striatum.
  • the forelimb test analyzes forelimb reaching and grasping and was performed pre- and post-lesion and post-transplantation. Animals were food deprived for 48 hours prior to the test, and tested daily for 5 days pre-lesion then twice after lesion at 3 week interval. Following grafting, the test is repeated 2 more times, at months 3 and 5. Animals were placed in a plexiglass chamber equipped with a double staircase. Food pellets are placed on 5 steps bilaterally for pre-graft testing, and unilaterally (on the affected limb side, contralateral to the rotation) after grafting. The animals are tested over a set time frame (e.g.
  • Substantia nigra pars compacta (SNpc) DA neurons in vivo exhibit an electrophysiological phenotype that differentiates them from most other neurons in the brain. In particular, they spike spontaneously at a slow (1-3 Hz) rate. Moreover, this slow spiking is accompanied by a slow, sub-threshold oscillatory potential. After 2-3 weeks in vitro, these same physiological features are displayed by SNpc DA neurons cultured from early postnatal mice.
  • the mDA neurons of the present inventions were tested for their electrical response signature.
  • Midbrain DA neurons of the present inventions on day 80-100 of culture were tested by single cell recording.
  • Electrophysiological measurements are contemplated for use in acute slice preparations, i.e. from biopsies of engrafted areas.
  • A9- versus A10 type graft-derived DA neurons will be identified in vivo based on testing for the autonomouse pacemaking activity that is specific to A9-type dopamine neurons that are most affected in PD. In other words, A10 type neurons do not have pacemaking activity
  • FIG. 26 Conditions were established for the in vivo recording of human pluripotent stem cell derived DA neurons in acute slice preparations, see, FIG. 26 .
  • grafted human DA neurons derived from pluripotent stem cells were measured for and discovered to have electrophysiological features typical of those seen in mouse substantia nigra pars compacta (SNpc), FIG. 26A where the top view shows reconstruction of a pacemaking neuron in the graft region.
  • Bottom shows an exemplary photomicrograph of a brain slice taken from the rat into which the hES-derived neurons were injected 9 months prior; the graft is outlined; a higher magnification image is shown inset at the bottom.
  • the slice was processed for tyrosine hydroxylase which shows up as white, FIG.
  • FIG. 26B shows an exemplary cell-attached patch recording from a putative DA neuron in the graft; Bottom shows an exemplary whole cell recording from the same cell. Recordings were made in the presence of glutamate and GABA receptor antagonists (50 ⁇ M AP5, 10 ⁇ M CNQX and 10 ⁇ M GABAzine) to eliminate synaptic input. These recordings demonstrated that the PS-derived neurons were autonomous pacemakers with normal intrasomatic voltage trajectories. Another neuron recorded in a graft sample had similar properties, FIG. 26C . For comparison, cell-attached and whole cell recordings from a dopaminergic neuron in SNpc of an adult mouse are shown.
  • GABA receptor antagonists 50 ⁇ M AP5, 10 ⁇ M CNQX and 10 ⁇ M GABAzine
  • CTx cortex
  • STr striatum
  • SNpc substantia nigra pars compacta
  • DA dopaminergic
  • This example described the discovery that large populations of midbrain DA neurons developed with characteristics of a PD patient's neurons when a PD patient's cell line, i.e. PINK1 mutant PD-iPSC cell, obtained in a manner that did not result in the destruction of an embryo, were used as the cell population for obtaining FOXA2/LIM1XA/TH+ DA neurons of the present inventions.
  • the inventors' contemplate isolating a starting cell population from a patient for use in the methods of making authentic DA neurons in vitro, where the patient has a symptom of Parkinson's disease (PD), for the potential advantage of using the treated cells in in vitro tests for 1) observing differentiation or functional abnormalities compared to authentic DA neurons from humans not having a neurological symptom, then 2) using an observed abnormality for developing a therapeutic treatment for reversing that abnormality and 3) treating the patient with the therapeutic treatment for reducing, i.e. reversing, a symptom of Parkinson's disease.
  • PD Parkinson's disease
  • the inventors' contemplate isolating a starting cell population from the same patient for deriving authentic DA neurons for use in transplantation treatment, where the patient has a symptom of Parkinson's disease (PD), for the potential advantage of reducing immunological rejection, i.e. transplantation rejection.
  • PD Parkinson's disease
  • reduction of transplantation rejection is contemplated by using a beginning cell source isolated from a human whose Major Histocompatibility Antigens (MHC) match (ie. Twin) or a human having an acceptable MHC tissue match for transplantation (such as a relative to the patient or an unrelated human expressing overlapping MHC molecules.
  • MHC Major Histocompatibility Antigens
  • a PINK1 Q456X mutant PD-iPSC line was differentiated using the novel floor-plate based midbrain DA neuron protocol (method) described herein which yielded midbrain DA neurons that expressed differentiation profiles comparable to those obtained from the novel floor-plate based midbrain DA neuron protocol differentiated H9 line. ( FIG. 20 ).
  • This example described the discovery that large populations of midbrain DA neurons developed with characteristics of a PD patient's neurons when a PD patient's cell line, i.e. PINK1 mutant PD-iPSC cell, obtained in a manner that did not result in the destruction of an embryo, were used as the cell population for obtaining FOXA2/LIM1XA/TH+ DA neurons of the present inventions.
  • PINK1 Q456X mutant PD-iPSC line was differentiated using the novel floor-plate based midbrain DA neuron protocol (method) of the present inventions which yielded midbrain differentiation profiles comparable to those obtained from the iPSC H9 line.
  • A-C Immunocytochemical analysis of PINK1 mutant PD-iPSC line at day 11 of differentiation (midbrain precursor stage) for FOXA2 (red), LMX1A (green) and DAPI (blue) (A), day 25 of differentiation (early postmitotic DA neuronal stage) for FOXA2 (red) and TH (green) (B) and for NURR1 (red) and TH (green) (C).
  • FIGS. 21-24 B. Genetic PD-iPSC Expressed a PD Like Phenotype of Protein Aggregation.
  • FIGS. 21-24 B. Genetic PD-iPSC Expressed a PD Like Phenotype of Protein Aggregation.
  • PINK1 mutant PD-iPSC showed evidence of ⁇ -synuclein (major component of Lewy body on PD patience) expression in cytosol of TH+ DA neurons at day 55 of differentiation using the novel floor-plate based midbrain DA neuron induction protocol, ( FIG. 21 a - b ).
  • ⁇ -synuclein positive cells also showed high expression of ubiquitin (classical Lewy body marker).
  • DA neurons derived from control iPS line showed expression of normal synaptic (as opposed to cytosolic) ⁇ -synuclein expression and very low levels of Ubiquitin ( FIG. 21 c - d ).
  • dimerized insoluble form of a-synulcein leads to aggregation in Lewy body.
  • the dimerized form of ⁇ -synuclein shows phospholylation of Serine 129 on ⁇ -synuclein.
  • PINK1 mutant PD-iPSC derived cells showed strong expression for Ser129 phosphorylated ⁇ -synuclein in contrast to control-iPSC derived cells that showed very low levels of expression ( FIG. 22 ).
  • PINK1 mutant PD-iPSC derived cells showed strong expression for Ser129 phosphorylated ⁇ -synuclein in contrast to control-iPSC derived cells that showed very low levels of expression.
  • floor-plate derived “authentic” midbrain DA neurons showed PD specific vulnerability and corresponding, specific, in vitro phenotypes.
  • DA neurons obtained using the classical MS5 stromal feeder based differentiation protocol yielded large numbers of TH+ neurons.
  • MS5 based TH+ cells were not authentic floorplate derived midbrain DA neurons. In cultures differentiated via the MS5 protocol, there were many ⁇ -synuclein positive cells. However, those cells did not co-express TH.
  • PD-iPSC derived TH+ DA neurons derived via floor-plate based protocol were more vulnerable to toxin challenge (valinomycin: mitochondria ionophore, 5 uM (ranging in concentration from 1-10 uM), 48 hr) than control-iPSC derived cells.
  • valinomycin mitochondria ionophore
  • 5 uM ranging in concentration from 1-10 uM
  • 48 hr mitochondria ionophore
  • TH+ neurons derived via the classic MS5 based protocol did not show differential vulnerability between PD- versus control-derived cells ( FIG. 24 ).
  • Entire cell viability assay with alamar-blue after 48 hrs of valinomycin treatment also showed differential cell survival in a specific dose range for toxin challenge (5 and 10 uM) when comparing PD-iPSC and control iPSC ( FIG. 25 ).
  • K-N low power images of immunocytochemistry for Tuj1 (red) and TH (green) by MS5 based protocol at day 60 of differentiation: PD-iPSC of normal (K), versus toxin challenge (L) conditions and control iPSC of normal (M), versus toxin challenge (N) conditions.
  • the descriptions herein show exemplary methods and uses for large-scale production of mDA neuronal cells resulting from differentiation by compositions and methods described herein.
  • the scalable generation i.e. methods contemplated to be successful for generating mDA neuronal cells from cultures containing a relatively small number of cells
  • PINK iPSC cells See, Table 9).
  • mice Using mouse cells, several genetic reporter strategies were proposed for use in identifying enrichment for neural cells in DA neuron transplantation paradigms (for example, identifying cells with expression of SOX1, Corin/Lmx1a, Ngn2, TH, Pitx or DAT). Functional testing was performed in primary cells from Ngn2-reporter mice (Thomposon et al., Exp Neurol. 198(1):183-98 (2006)) and from Lmx1A-reporter mice also sorted for Corin (Jönsson, Exp Neurol. 219(1):341-54 (2009). For mouse ESC derived populations studies were performed using SOX1 (Barraud et al., Eur J. Neurosci.
  • mice ESC reporter lines mouse ESC reporter lines. Additionally, during the development of the present inventions, the inventors performed a comprehensive transplantation study directly comparing the in vivo performance of three purified mouse ESC-derived populations representing sequential stages of DA neuron development: Midbrain precursors (Hes5::GFP), early postmitotic cells (Nurr1::GFP), and mature DA neurons (Pitx3::YFP).
  • Nurr1-expressing DA developmental as particularly suitable for grafting and demonstrated that purified DA neurons are capable of efficient engraftment in vivo. Furthermore, these results were used for selection of cells at day 25 of differentiation (onset of Nurr1 expression) for use in grafting hESC-DA neurons into mouse, rat and rhesus monkey models of PD.
  • FACS sorting may be problematic for establishing clinical grade DA neuron master cell banks (i.e. developing frozen stocks of human DA neurons for use in transplantation) given the length of time that would be required for sorting batches of the approximately 10 9 cells required for each transplant in addition to potentially high costs and lower cell yield after recovery from storage.
  • DA neurons based on surface marker expression In contrast to genetic reporter systems and FACS based cell isolation, the inventors contemplated isolation of DA neurons based on surface marker expression using alternative strategies for cell separation techniques contemplated for use in PD patients.
  • magnetic bead sorting e.g. CliniMACS® system
  • CliniMACS® system was widely used in FDA-approved, cell-based applications and allowed for rapid and cost-effective isolation of up to 10 10 cells under GMP-compliant conditions, i.e. conditions approved for isolating cells for use in humans.
  • magnetic bead sorting is contemplated for enrichment of mature DA neurons, for example, using CD142 attached to magnetic beads for enriching Nurr1+ neurons for use in grafts.
  • Cell surface marker expression data collected during the development of the present inventions showed identification of several novel cell surface markers expressed on midbrain DA neurons. Specifically, markers for further identifying cells, such as specific cells that would mature into DA neurons, mature DA neurons of the present inventions and A9 cells, were found.
  • Two main strategies were used to identify such surface markers: first, an unbiased gene expression screen in genetic reporter lines ( FIG. 27 a ) showed several candidate markers, including CD142 and a marker termed DCSM1, that was selectively expressed in midbrain DA neurons and appeared to specifically mark A9-type DA neurons ( FIG. 27 b .
  • a second strategy was the use of a CD cell surface marker screen in hESC derived DA neurons which tested for 242 commercially available antibodies in 96 well format ( FIG.
  • a CD surface marker screen for WA09-derived DA neurons at day 25 of differentiation tested for up to 242 individual antibodies. These results were compared to duplicate screens of a broad range of other WA09 derived neural cell types (e.g. hESC-derived HB9::GFP+ motoneurons, hESC derived cortical neurons, hESC derived Nkx2.1::GFP+ ventral forebrain precursors, and several other hESC derived neuron types.
  • the resulting database of surface marker expression profile was then used to select candidate CD markers selectively enriched in any given subtype such as midbrain DA neurons ( FIG. 27 ).
  • One of the markers discovered associated with hESC DA neuron differentiation was CD142.
  • CD142 is expressed before Nurr1+.
  • a midbrain DA neuronal cell population sorted for CD142 has Nurr1+ and Nurr1 ⁇ cells.
  • a midbrain DA neuronal cell population sorted for CD142 has Nurr1 ⁇ cells.
  • cultured Nurr1 ⁇ CD142+ sorted midbrain DA neuronal cell population begin expressing Nurr1 (i.e. become Nurr1+) in up to two days after sorting.
  • CD63 and CD99 were markers enriched on hESC derived DA neurons.
  • DA neuronal cultures are enriched for DA neurons by sorting or selecting from markers including but not limited to CD142, CD63, CD99, DCSM1, Nurr1+, etc.
  • CD142 typically marks approximately 30% of the total cell population at day 25 of differentiation ( FIG. 28 a ). Selectivity of CD142 for Nurr1+ DA neuron stage was confirmed in multiple independent hESC and hiPSC lines ( FIG. 28 b ).
  • CD142 selectively depletes other neuron subtypes such as GABA and Serotonergic neurons. ( FIG. 28 c - f ).
  • A9 derived vs. A10 derived DA neurons were found to have distinct in vitro and in vivo functional properties and innervations patterns specific to their role in mesostriatal versus mesolimbic function.
  • the authentic mDA neurons produced by methods of the present inventions gave rise to neurons with having more A9 than A10 characteristics.
  • authentic mDA neurons that were TH+ at least in part expressed Girk2, a marker used to define A9 type DA neurons.
  • Many mature DA neurons exhibited autonomous pacemaking activity that is a functional feature present in A9 but not A10 type DA neurons.
  • some TH+ cells generated in vitro were not of A9 identity.
  • A9 type neurode versus A10 neurons.
  • the inventors contemplated enrichment procedures such as those described herein, for providing purified populations of human A9 type authentic mDA neurons (versus A10) neurons.
  • the inventors discovered at least two markers unique to A9 type neurons and at least two markers at unique to at least A10 type neurons.
  • A9 type neurons are identified by (Girk2, Aldh1) versus A10 (Calbindin, Otx2) markers.
  • Candidate markers were obtained from a gene expression screen, such as described herein, and candidate CD-antibodies from a surface marker screen, as described herein.
  • Populations In another method, global transcriptome analysis in purified populations of mouse ESC derived mDA neurons at distinct stages of differentiation (using BAC transgenic technology; see FIG. 27 a,b ). Surface markers were discovered in a surface marker profile on DA neurons derived from WA09 RCB with the following exemplary methods. RCB WA09-derived DA neurons at day 25 of differentiation were dissociated and replated onto 96 well plates, followed by exposure to the 242 CD antibodies and data analysis using the Operetta high content scanner.
  • DA-enrichment was tested for at least 5 additional antibodies which bound to CD markers identified in these screens (for examples, CD142, CD63 and CD99).
  • CD markers identified in these screens for examples, CD142, CD63 and CD99.
  • Candidate CD-positive versus CD-negative cells were assessed using the DA QC assays, including expression of FOXA2/TH and TH/Nurr1 (see, Table 7).
  • global gene expression profiles are contemplated for comparison of unsorted to CD142+ cells.
  • cells sorted/separated expression a desired marker were used in short-term and long-term in vivo studies as described herein.
  • DCSM1 DA cell surface marker 1).
  • A9 DA neurons exhibited specific functional features as described herein for hESC derived A9 neurons.
  • In vivo studies were performed for cells expressing DCSM1 and other markers in order to confirm i) A9 marker expression (Girk2, Aldh1) versus A10 (Calbindin, Otx2), ii) graft DA fiber outgrowth and iii) electrophysiological A9 properties in slice preparation of the grafted cells (see FIG. 26 ).
  • graft integration and extent of DA fiber outgrowth are challenges in grafting methods including the fetal grafting studies in PD patients.
  • One problem encountered with graft tissues and cells is limited fiber outgrowth from these grafts when treating patients. This problem is particularly critical in humans since patient recovery requires extensive striatal reinnervation.
  • achieving adequate reinnervation after a tissue graft required multiple injections of cell deposits across the striatum. Each injection can cause striatal damage and inflammation along with other surgical risk.
  • Such risks include the injury of a blood vessel during cell injection that could potentially induce a stroke or seizures in the patient.
  • PSA is a natural cell surface sialic acid homopolymer (i.e.
  • PSA polysialyltransferase
  • NCAM neural cell adhesion molecule
  • CD56 NCAM
  • PSA appeared to function in regulating plasticity of some cell behaviors that required changes in cell-cell interactions, including cell migration and axon outgrowth. While highly expressed in the embryo, PSA was down regulated in adult tissues with the exception of localized regions of the CNS that maintain structural and physiological plasticity (such as hippocampus, suprachiasmatic nucleus, SVZ).
  • polysialic acid was contemplated for use in promoting fiber outgrowth of engrafted cells.
  • PSA polysialic acid
  • Examples of PSA use in other cell types are described in WO 2006/042105 herein incorporated by reference in its entirety.
  • the inventors contemplate the use of authentic DA neurons in combination with PSA as described herein.
  • PSA polysialic acid
  • a side-by-side comparison of PSA-enhanced versus control treated ES-derived DA neuron grafts showed behavioral recovery in the PSA group (when grafts were based on transplantation of 55,000 cells each) but not observed in control cells.
  • Transplantation of 100,000 ES derived DA neurons in the mouse brain showed behavioral recovery in both PSA-treated and control-treated ES-derived DA neurons suggesting that grafts derived from 100,000 cells are sufficient to reinnervate the mouse brain without PSA enhancement.
  • engineered expression of PSA expression on the surface of authentic DA neurons is contemplated for use in procedures for treatment of patients with PD.
  • PSA When PSA was induced on neuronal cells of the present inventions it was identical to the PSA polymer that occurred naturally in brain cells thus unlike the use of other cell surface molecules for engineering therapeutic cell types, cells engineered for PSA expression are contemplated to have little antigenicity in vivo when used on cells for engraftment procedures in humans. Moreover, high PSA levels on engrafted cells, either endogenouse neural precursors (Battista et al., J. Neurosci. 30(11):3995-4003 (2010)), Schwann cells (Ghosh et al., Glia.
  • PSA expression is contemplated on neuronal cells for use in engraftment procedures.
  • PSA is contemplated for use in preparing cells for therapeutic use by overcoming problems encountered when using other types of induced cell surface marker expression.
  • the use of PSA expression was reproducible in protocols that cross vertebrate species (such as using mouse PST genes expressed in human cells) because its acceptors are also consistent in structure across species and are found on the majority of cell surfaces.
  • hPST human polysialyl-transferase
  • At least 3 clones that retain high, uniform levels of PSA-NCAM during differentiation and perform well in the QC parameters will advance to assessment of the neurite outgrowth in PST-overexpressing hESC-derived DA neurons
  • Selected control and PST-overexpressing hESC clones were differentiated into DA neurons using the standard protocol described herein, followed by cell fixation and analysis at days 25 and 50.
  • the number and length of TH-positive fibers in such cultures were quantified with the Operetta High Content Microscope.
  • PST-overexpressing and control hESC clones that advance from in vitro studies above, were differentiated again into DA neurons and transplanted into a rat model of PD. Short-term grafts (4-6 weeks) to determine survival, PSA-NCAM expression and neurite outgrowth were done. For each clone that passed short-term in vivo parameters were subjected to long-term grafting studies. For those studies animals received half or a quarter of the standard (200 ⁇ 10 3 ) dose of cells. These studies were to address whether increased PSA leads to increased long-term survival after transplantation (5 months), and whether smaller DA neuron numbers are capable of matching or outperforming the functional capacity of non-PST grafts transplanted at standard cell doses ( FIG. 27 ).
  • the following example shows enhancement of polysialic acid expression that improved the function of ES-derived dopamine neuron grafts in Parkinsonian mice.
  • ES cells expressing GFP under control of Nurr1 promoter were stably transduced with a lentiviral vector ubiquitously expressing polysialyltransferase (PST).
  • PST polysialyltransferase
  • Transduced cells showed a dramatic increase in PST mRNA as compared to controls ( FIG. 30A ).
  • Expression of PST was observed to be sufficient for PSA synthesis on NCAM. Accordingly, PSA-NCAM expression was greatly increased in PST-modified cells at day 14 of DA neuron differentiation ( FIG. 30B-E ). Both the endogenous and induced cell surface PSA on ES-derived DA neurons could be removed ( FIG.
  • PSA enhancement provided a significant augmentation of the ability of grafted DA neurons to innervate host striatum and attenuate PD functional deficits. Therefore clinical translation is contemplated comprising DA neurons of the present inventions for providing cells prior to transplantation.
  • the cells will be genetically manipulated for expressing PSA.
  • PST may be delivered directly to the cells via exposure to the purified enzyme and substrate, in vitro, prior to transplantation.
  • PSA strategy for human translation in PD grafting is contemplated to minimize the need for multiple injections and thereby reduce the surgical risks resulting from these multiple injections.
  • this technology is contemplated for use on other cell types and species, for example, augmenting the migration of grafted Schwann cells in creating a bridge (for example, cell-cell communication) for re-growth of axons at the site of spinal cord injury.
  • a bridge for example, cell-cell communication
  • mice Six-week old 129S3/SvImJ mice (Jackson Laboratory) were kept under controlled temperature with food and water available ad libitum. Experimental procedures were performed according to NIH and institutional animal use guidelines and approved by the local Institutional Animal Care and Use Committee (IACUC) and the Institutional Biosafety Committee (IBC).
  • IACUC Institutional Animal Care and Use Committee
  • IBC Institutional Biosafety Committee
  • 6OHDA injection and amphetamine-induced test Animals were anesthetized with sodium pentobarbital (10 mg/kg) and injected in the right striatum with 2 ⁇ l of 6OHDA (4 ⁇ g/ ⁇ l in saline, 0.5% ascorbic acid). The injections were performed with a Hamilton syringe at coordinates: 0.5 mm posterior, 1.8 mm lateral relative to bregma and 2.5 mm ventral to brain surface. Before the surgery animals received a single i.p. injection of desipramine (25 mg/Kg, Sigma). Two weeks after surgery animals were scored in the amphetamine-induced rotation test.
  • a Nurr1::GFP BAC transgenic BAC mouse ES reporter cell line i.e., GFP expression is driven by Nurr1 promoter
  • a lentivirus pLenti, Invitrogen
  • ES cells were propagated on mitomycin C-treated MEFs (StemCell Technologies) in DMEM (Invitrogen), 10% FBS (HyClone) supplemented with 1,400 units/ml LIF (ESGRO; Invitrogen), 2 mM L-glutamine, 1 mM ⁇ -mercaptoethanol, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin (Invitrogen).
  • DA differentiation was induced according to Barberi et al., Nat Biotechnol 21, 1200-1207 (2003), with modifications. Briefly, cells were differentiated on MS5 feeder cells in gelatin-coated dishes (10,000 cells/10 cm dish) and cultured for four days on serum replacement media (SRM).
  • SRM serum replacement media
  • Sonic hedgehog SHH, 200 ng/ml
  • FGF8 100 ng/ml
  • the media was changed to N2 supplemented with SHH, FGF8 and bFGF (10 ng/ml).
  • terminal differentiation was induced by withdrawal of SHH, FGF8 and bFGF and the addition of ascorbic acid (AA, 200 ⁇ M) and BDNF (20 ng/ml).
  • Nurr1::GFP sorted cells were analyzed for viability and resuspended in N2 with BDN and AA to a final concentration of 55,000 cells/ ⁇ l
  • One ⁇ l was injected into the lesioned mouse striatum with a 50 ⁇ m tipped fine glass capillary at coordinates: 0.3 mm posterior, 1.5 mm lateral from bregma and 2.2 mm ventral to the brain surface.
  • An aliquot of the cell suspension was re-plated in matrigel-coated 6 mm dishes for further characterization.
  • cells were fixed with paraformaldehyde for 10 min at 40 C, washed twice with PBS, blocked with 5% BSA (0.1% Triton X-100 in PBS) and incubated with primary antibodies for 2 hrs at room temperature: rabbit anti-GFP (1:1000, Invitrogen), mouse IgM anti-PSA (1:2000, 5A5), mouse anti-NeuN (1:800, Chemicon), mouse anti-TH (1:1000, Sigma), goat anti-FoxA2 (1:800, Santa Cruz), goat anti-Engrailed (1:800, Santa Cruz). Cells were then incubated with Cy-conjugated secondary antibodies (1:1000, Jackson).
  • EndoN treatment To remove PSA from NCAM, the night before harvesting, cells were treated with 20 units of endoN, a phage enzyme that specifically removes PSA 7-9. Cells were then harvested and injected as described before but were resuspended in N2 with BDNF and AA and 5 units of endoN. We previously assessed that the injection of the same amount of endoN alone into lesioned mice did not improve animal behavior.
  • PST mRNA and PSA-NCAM analysis in vitro were treated with WB buffer (PBS with 1% NP40, 150 mM NaCl, 1 mM EDTA, and 1 ⁇ protease/phosphatase inhibitors added immediately before extraction, at pH of 7.4) and sonicated twice for 5 sec, centrifuged and resuspended in Laemli buffer (LB). Aliquots without LB were saved for protein determination. Equal amounts of protein were loaded into 6% sodium dodecyl sulfate-polyacrylamide gel electrophoresis gel (BioRad). Proteins were transferred by electrophoresis onto polyvinylidene membranes (Millipore).
  • WB buffer PBS with 1% NP40, 150 mM NaCl, 1 mM EDTA, and 1 ⁇ protease/phosphatase inhibitors added immediately before extraction, at pH of 7.4
  • LB Laemli buffer
  • the membranes were blocked for 1-6 hr in 0.1% Triton X-100 TBS (TBS-T) with 5% non-fat dry milk and incubated overnight with anti-NCAM antibody (1:10,000, Santa Cruz) in TBS-T with 5% milk. Blots were then incubated with peroxidase-conjugated secondary antibody (1:10,000, Jackson) and detected with ECL detection method (Amersham Pharmacia Biotech). Protein levels were quantified using ImageJ software.
  • PSA-NCAM FACS analysis cells were harvested with accutase treatment for 45 min, washed once and incubated with mouse IgM anti-PSA (1:250, 5A5) for 25 min on ice, washed once with N2 media and incubated with Cy3-conjugated anti-mouse-IgM (1:250, Jackson) for another 25 mM on ice. Cells were washed once with N2 and resuspended with 0.1% BSA with 7AAD and analyzed in a FACS Calibur cell sorter. As control, no primary antibody was added.
  • Immunohistological and stereological procedures Free floating coronal sections were blocked in 0.1% Triton X-100, 5% donkey serum in PBS for 30 mM at room temperature and incubated 48 hrs at 4° C. with different antibodies: rabbit anti-GFP (1:300), chicken anti-GFP (1:200, Chemicon), mouse anti-TH (1:200), mouse IgM anti-PSA (1:1000), mouse anti-NeuN (1:400), goat anti-FoxA2 (1:300), rabbit anti-Girk2 (1:300, Alomone Labs), mouse anti-synapsin (1:200, BD Transduction Laboratories).
  • the number GFP+ and TH+ cells was counted in one-in-three sections encompassing the whole brain under a 40 ⁇ objective, and the total number of cells/graft estimated. Double-labeled cells were analyzed in single optical planes through the entire z-axis.
  • GFP+ cells were analyzed for each marker.
  • process outgrowth analysis confocal z-scans were performed at 0.8 ⁇ m intervals through the entire z-axis (20-40 ⁇ m) with a pinhole of 1 ⁇ m under a 40 ⁇ objective. Sections were scanned from the injection site laterally until no processes were observed. 3-D projections encompassing the whole scanned area were sequentially matched.
  • GFP and TH intensity analysis the entire scanned area was divided into five successive 100 ⁇ m zones away from the transplant and the intensities were measured using ImageJ software. Data were normalized to the intensity in the zone nearest the graft (zone I) to control for any potential differences in graft size.
  • Lesioned animal groups received one of 3 doses of wild type cells or cells expressing PST or pretreated with PST enzyme. They were processed for behavioral testing following the paradigms discussed in Table 11 (amphetamine rotations, cylinder test, stepping test). The doses chosen (for examples, 200 k, 100 k, 50 k cells) were used successfully in mice such that resulting neurons showed results as described herein for mouse PST genes.
  • a nontransgenic method of PSA induction Pre-treatment of cells with the PST enzyme which results in polysialylation and increased expression of surface PSA.
  • the purified Neisseria meningitides ⁇ -2,8-polysialyltransferase (PSTnm) operated in an extracellular environment when using a commercially available non-toxic substrate (i.e. CMP-sialic acid) and produced a polymer chemically identical to mammalian PSA.
  • a commercially available non-toxic substrate i.e. CMP-sialic acid
  • an active fragment of this enzyme was effective for adding PSA to therapeutic proteins in vitro to augment in vivo pharmacokinetics.
  • PSA was synthesized by PSTnm directly on surfaces of a wide variety of cell types in vitro, including mouse and human ESCs ( FIG. 35A-E ).
  • PSA expression via PSTnm occurred in less than an hour which overcame the slow PST transgene induction.
  • GMP-grade reagents are used in these procedures for meeting clinical use protocols, and having a transient nature of biochemically-generated PSA expression matching the expected time frame for DA fibers leaving the graft core and entering the host brain, necessary for avoiding some dangerous side effects of using cells engineered for engraftment.
  • the following example shows enzymatic engineering of PSA on hESC-derived DA neurons using the purified bacterial polysialyltransferase, PSTnm, to enhance transplant efficacy.
  • DA neurons externally treated with PSTnm is contemplated for use in the producing cells for engraftment.
  • Both mammalian PST and PSTnm produced chemically identical chains of PSA.
  • Increased PSA on hESC-derived DA neurons should persist for several weeks, sufficient for DA fibers to exit graft core. Because PSTnm is removed prior to grafting, immunogenicity to this enzyme contaminating the grafted cells should not be factor.
  • PSTnm was produced from an engineered fragment with enhanced solubility and activity characteristics (Willis et al., Characterization of the alpha-2,8-polysialyltransferase from Neisseria meningitidis with synthetic acceptors, and the development of a self-priming polysialyltransferase fusion enzyme. Glycobiology 18, 177-186 (2008)). Cultures of hESC were induced to differentiate into DA neurons before PSTnm exposure, exposure to substrate or both. Cultures were examined at different time-points of exposure (10 min to 6 hrs) by quantitative immunofluorescence (Operetta) and western blotting to determine the speed and levels of polysialylation.
  • Day 25 differentiated hESC-derived DA neurons will be incubated with the optimum concentrations of PSTnm and substrate using the conditions described herein.
  • PSA+mDA neurons will be transplanted in short- and long-term assays as described herein and in FIG. 29 .
  • enhancing PSA expression on Schwann cells used for engraftment resulted in enhancement of Schwann cell migration and axonal growth which further resulted in dramatic effects on increasing locomotor function ( FIG. 30A-D ).
  • increasing PSA expression on Schwann cells in vitro is contemplated for use in engraftment procedures in humans having spinal cord injury.
  • hESC-based cell therapy procedures One of several concerns for hESC-based cell therapy procedures is the possibility of introducing contaminating undifferentiated cells that resisted differentiation which under post engraftment conditions develop into cells that cause harm to the patient. In the case of a pluripotent cell, one harmful result is teratoma formation that endangers the patient's life. Teratoma formation using hESC derived cells was reported following short-term neural differentiation protocols based on spontaneous cell differentiation.
  • ES derived neural cell types unlike their mouse ESC-derived counterparts, rarely resulted in teratoma formation following appropriate neural differentiation strategies as described in the current invention (i.e. monolayer culture, dual-SMAD-inhibition protocol and growth in cytokines that do not promote proliferation).
  • appropriate neural differentiation strategies i.e. monolayer culture, dual-SMAD-inhibition protocol and growth in cytokines that do not promote proliferation.
  • teratoma formation was not observed.
  • teratomas were not observed in PD transplantation procedures of the present inventions using human cells for grafts. The difference between the use of human vs.
  • mouse cells for engraftment procedures for use in humans is contemplated to be related to the different stage of pluripotency captured in human versus mouse ESCs, whereby human cells are thought to match the properties of a pluripotent stage described as Epi-SCs, unlike mouse ESCs which may be at a different developmental stage.
  • hESC derived neuroepithelial structures i.e. neural rosette-type
  • This in vivo expansion of grafted neuroepithelial cells was observed in various neural transplantation paradigms including hESC derived DA neuron transplantation studies in rodent Parkinson's and Huntington's disease models.
  • Those “neural rosette-type” proliferating cells represented non-transformed primary cells with a high intrinsic growth potential which resulted in large grafts composed of ectopic, mostly cortical-type tissue in grafted animals.
  • several strategies were used to eliminate contaminating pluripotent or neuroepithelial cells at the time of grafting (e.g. selection for SSEA-4 (pluripotent marker) or Forse-1 (neuroepithelial marker). These strategies were partially successful when using rosette-based differentiation strategies and neural overgrowth was still observed in a subset of grafts sorted for Forse1 or sorted negatively for SSEA-4.
  • preventing “hot spots” of neuronal clusters secreting L-Dopa and other compounds are major advantages of using the methods of the present inventions over other methods for providing cells for use in engraftment procedures. Thus use of the methods of the present inventions is contemplated to minimize risk to patients.
  • human ESCs are contemplated for use in methods for making and using cells for engraftment procedures in humans, in other words, cell therapy for the treatment of PD.
  • human ESCs have numerous advantages over using human iPSCs in methods of the present inventions, such as, for one example, for use in providing engraftable midbrain DA neurons for use as a PD cell therapy.
  • iPSCs induced pluripotent stem cells
  • the use of induced pluripotent stem cells (iPSCs) as a cell source for DA neuron derivation has several advantages, such as providing a genetically matched cell source for each patient.
  • iPSCs induced pluripotent stem cells
  • hESCs were observed to acquire mutations over time in culture the timing and rate of such mutations appeared to differ from hiPSCs, where hESCs were generally considered more genetically stable than iPSCs, for examples, see, Hussein, et al., Nature 471, 58-62 (2011); Mayshar, et al. Cell Stem Cell 7, 521-531 (2010); Lister, et al. Nature 471, 68-73 (2011); Laurent, et al. Cell Stem Cell 8, 106-118 (2011). Additionally, standard operating procedures were devised for several hESC lines that satisfied the rigorous safety tests required by the FDA for cellular and gene therapy products. The FDA has approved two groups in the United States to advance hESC-based cellular therapies to clinical use.
  • Geron Corporation entered a Phase I trial with hESC-derived oligodendrocyte precursor cells (GRNOPC1), and Advanced Cell Technology (ACT), Inc. has two current Phase I/II trials using hESC-derived retinal pigmented epithelial cells to treat Stargardt's Macular Dystrophy (trial NCT01345006) and Advanced Dry Age Related Macular Degeneration (trial NCT01344993).
  • GRNOPC1 hESC-derived oligodendrocyte precursor cells
  • ACT Advanced Cell Technology
  • both of the FDA-approved, hESC-based clinical trials target nervous system disorders show advantages of using hESC-based methods for providing engraftment material for treating nervous system diseases and injuries.
  • the nervous system is considered an immuno-privileged site since foreign tissue (allografts) elicits weak immune responses when compared to the same graft placed into the periphery.
  • foreign tissue allografts
  • after twenty-five years of transplanting fetal cells into human brains it was found that some allogenic neurons survived for up to 16 years in the human brain with transient immunosuppression. Therefore it appears that identical antigenic matching between a cell source and the graft recipient is not essential.
  • hESCs are contemplated as a universal, allogenic source of DA neurons for treating PD in addition to other nervous system diseases, disorders and injuries.
  • Patients receiving cells of the present inventions are contemplated to have a clinical diagnosis of PD.
  • Authentic DA neurons grafts are contemplated for use in early intervention and moderate-to-severe PD, including patients in whom there is insufficient symptomatic control with available medications, such as levodopa, adjunctive medication, etc.
  • patients contemplated to receive neuron grafts have subtle signs in early PD (e.g. by the use of neuroimaging for detecting dopaminergic deficits, FDG-PET, and signs of dyskinesias, etc.
  • Dyskinesia as measured by the Unified Dyskinesia Rating scale (UDysRS) (Goetz, et al., Mov Disord. 23, 2398-2403 (2008)) is contemplated for use in monitoring patients before and after engraftment. Patients may also have “scans without evidence of dopaminergic deficits” (SWEDDS), some of whom may have dystonia or essential tremor. Brain MRI would be done in order to identify patients with other (non dopa) contributory factors to Parkinsonism. In some embodiments, patients would have a positive response to levodopa.
  • UDysRS Unified Dyskinesia Rating scale
  • Determining pre- and post-transplantation parameters and endpoints for subject monitoring such as motor evaluation, non-motor evaluation, quality of life, and also the use of neuroimaging and other biomarkers.
  • Motor function The UPDRS and newly-validated MDS-UPDRS (Goetz, et al., Mov Disord. 23, 2129-2170 (2008)) are widely used for measuring PD motor symptoms. However, other tests including 10 m walk or 6 minute walk tests, timed up and go, functional gait assessment, functional reach, and others are contemplated for more patient-oriented outcome measures.
  • Non-motor function Measures will primarily target cognitive, psychiatric outcomes and dysautonomia in addition to addressing cognition, depression, anxiety, apathy, sleep, fatigue, psychosis, and other non-motor symptoms before and after engraftment.
  • Quality of life PD-specific questionnaires (PD-QUALIF) and/or well-validated quality of life scales such as the SF-36 are contemplated to monitor patient outcomes.
  • Neuroimaging and other biomarkers Functional imaging was widely used in surgical PD trials. While dopamine-based imaging (such as FDOPA-PET) is contemplated for use in examining graft maintenance, neuroimaging techniques using other ligands are contemplated for use including imaging-based markers, for example targeting inflammation, and of non-imaging systemic markers as exploratory data collection. Imaging would find use in pre-operative planning, e.g. extent and location of DA depletion within the basal ganglia and incorporation of PET data in tailoring surgical planning for each patient. Location of graft placement and number of cell deposits. In some embodiments, the putamen (Freed, et al. N. Engl. J. Med.
  • MRI with a Clearpoint system which provides real time imaging and visualization of the trajectory path and targeting accuracy is contemplated for monitoring the engrafted cells. This system is used in placement of Deep Brain Stimulation electrodes in PD patients. Cell number and Composition of the graft. Fetal trials were performed with essentially unknown number of DA neurons since they used fetal graft material with numerous cell types. Based upon data provided herein, an estimated 100,000-200,000 surviving TH+ neurons are contemplated for recovery of DA neuronal function.
  • immunosuppression of grafted patients is contemplated, at least 6 months up to the lifetime of a patient. In some embodiments, patients will not be immunosuppressed for purposes of having engrafted tissue.
  • TABLE 6 shows an exemplary list of antibodies used as markers, including concentration (i.e. dilution) of antibodies used and exemplary sources of antibodies.
  • bound antibodies were identified with any of Alexa488, Alexa555 and Alexa647-conjugated secondary antibodies (Molecular Probes, Carlsbad, California).
  • biotinylated secondary antibodies were used to identify the bound primary antibodies followed by visualization via DAB (3,3′-Diaminobenzidine) chromogen.
  • Antibody primary Dilution Source Location Human nuclear 1:100 Millipore Billerica, MA antigen Human cell 1:100 Santa Cruz Santa Cruz, CA adhesion molecule Tyrosine 1:1000/1:500 Pel-Freez/ Rogers, Hydroxylase (TH) ImmunoStar AR/Hudson, WI ⁇ -tubulin III 1:500/1:2000 Covance Littleton, CO/ Princeton, NJ Doublecortin 1:100 Millipore Billerica, MA Human specific 1:300 R&D Minneapolis, MN Nestin Nestin # 130 1:50 R.
  • Human ESC (H9, H1) and iPSC lines (2C6 and SeV6) were subjected to a modified Dual SMAD-inhibition (Chambers, et al. Nat. Biotechnol. 27:275-280 (2009), herein incorporated by reference) based floor plate induction (Fasano, et al., Cell Stein Cell 6:336-347 (2010), herein incorporated by reference) protocol. Exposure to SHH C25II, Purmorphamine, FGF8 and CHIR99021 were optimized for midbrain floor plate and yield of novel populations of DA neuron (see FIG. 1 d ).
  • DA neuron survival and maturation factors Perrier, et al. Proc Natl Acad Sci USA 101:12543-8 (2004), herein incorporated by reference
  • AA AA
  • BDNF BDNF
  • GDNF GDNF
  • TGF ⁇ TGF ⁇
  • dbcAMP dbcAMP
  • DA neurons were injected stereotactically in the striata of the animals (150 ⁇ 10 3 cells in mice, 250 ⁇ 10 3 cells in rats) and a total of 7.5 ⁇ 10 6 cells (distributed in 6 tracts; 3 on each side of brain) in monkeys.
  • Behavioral assays were performed at monthly intervals post-grafting, including amphetamine mediated rotational analysis as well as a test for focal akinesia (“stepping test”) and limb use (cylinder test). Rats and mice were sacrificed at 18-20 weeks and the primates at 1 month post grafting. Characterization of the grafts was performed via stereological analyses of cell number and graft volumes as well as a comprehensive phenotypic characterization via immunohistochemistry. Culture of undifferentiated human ES cells.
  • hESC lines H9 WA-09, XX, passages 27-55 from when 10/2009
  • H1 WA-01, XY, passages 30-40 from when 6/2010
  • iPS cell lines 2C6 Kim, et al. Cell Stem Cell 8:695-706 (2011), herein incorporated by reference
  • XY, passages 20-30 XY, passages 20-30
  • SeV6 XY, passages 20-30; derived from MRC-5 embryonic fibroblasts using non-integrating 4 factor Sendai vector system (Ban, et al. Proc. Natl. Acad. Sci.
  • LDN-193189 100 nM (ranging in concentration from 0.5-50 ⁇ M, Stemgent, Cambridge, Mass.), SB431542 (10 ⁇ M (ranging in concentration from 0.5-50 ⁇ M, Tocris, Ellisville, Mich.), SHH C25II (100 ng/ml (ranging in concentration from 10-2000 ng/ml, R&D, Minneapolis, Minn.), Purmorphamine (2 ⁇ M (ranging in concentration from 10-500 ng/ml, Stemgent), FGF8 (100 ng/ml (ranging in concentration from 10-500 ng/ml, R&D) and CHIR99021 (CHIR; 3 ⁇ M (ranging in concentration from 0.1-10 ⁇ M, Stemgent).
  • SHH treatment refers to exposure, i.e. contact, of cells to a combination of SHH C25II 100 ng/ml+Purmorphamine (2 ⁇ M).
  • Cells were plated (35 ⁇ 40 ⁇ 10 3 cells/cm 2 ) and cultured for 11 days on matrigel or geltrex (used as purchased) (BD, Franklin Lakes, N.J.) in Knockout serum replacement medium (KSR) containing DMEM, 15% knockout serum replacement, 2 mM L-glutamine and 10- ⁇ M (ranging in concentration from 1-25 ⁇ M ⁇ -mercaptoethanol.
  • KSR Knockout serum replacement medium
  • KSR medium was gradually shifted to N2 medium starting on day 5 of differentiation, by mixing in ratios of 75% (KSR):25% (N2) on day 5-6, 50% (KSR):50% (N2) day 7-8 and 25% (KSR):75% (N2) on day 9-10, as described previously (Chambers, et al. Nat. Biotechnol. 27:275-280 (2009), herein incorporated by reference).
  • media was changed to Neurobasal medium/B27medium (1:50 dilution)/L-Glut (effective ranges 0.2-2 mM)) containing medium (NB/B27; Invitrogen) supplemented with CHIR (until day 13) and with BDNF (brain-derived neurotrophic factor, 20 ng/ml ranging from 5 to 100; R&D), ascorbic acid (AA; 0.2 mM (ranging in concentration from 0.01-1 mM), Sigma, St Louis, Mich.), GDNF (glial cell line-derived neurotrophic factor, 20 ng/ml (ranging in concentration from 1-200 ng/ml); R&D), TGF ⁇ 3 (transforming growth factor type ⁇ 3, 1 ng/ml (ranging in concentration from 0.1-25 ng/ml); R&D), dibutyryl cAMP (0.5 mM (ranging in concentration from _0.05-2 mM); Sigma), and DAPT (10 nM (ranging in concentration from 0.5-50 nM); Tocris, CH
  • cells were dissociated using Accutase® (Innovative Cell Technology, San Diego, Calif.) and replated under high cell density conditions (for example from 300-400 k cells/cm 2 ) on dishes pre-coated with polyornithine (PO); 15 ⁇ g/ml (ranging in concentration from 1-50 ⁇ g/ml)/Laminin (1 ⁇ g/ml) (ranging in concentration from 0.1-10 ⁇ g/ml)/Fibronectin (2 ⁇ g/ml (ranging in concentration from 0.1-20 ⁇ g/ml) in differentiation medium (NB/B27+BDNF, AA, GDNF, dbcAMP (ranging in concentration as described herein), TGF ⁇ 3 and DAPT (ranging in concentration as described herein) until the desired maturation stage for a given experiment.
  • Accutase® Innovative Cell Technology, San Diego, Calif.
  • hESCs were induced towards neural fate by coculture with irradiated MS5 cells in KSR supplemented with SB431542 and Noggin (250 ng/ml (ranging in concentration from 10-1000 ng/ml); R&D), from day 2-8 and SHH+FGF8 from day 6-11 of differentiation.
  • neural rosettes were manually isolated and cultured (P1 stage) in N2 medium supplemented with SHH, FGF8, BDNF and AA as described previously (Perrier, et al. Proc Natl Acad Sci USA 101:12543-8 (2004), herein incorporated by reference). After 5-7 days in P1 stage, rosettes were again harvested mechanically and triturated following incubation in Ca 2 /Mg 2 -free Hanks' balanced salt solution (HBSS) for 1 h and replated on polyornithine (PO)/Laminin/Fibronectin coated plates.
  • HBSS Hanks' balanced salt solution
  • Patterning with SHH/FGF8 was continued for 7 days at P2 stage followed by final differentiation in the presence of BDNF, AA, GDNF, TGFb3 and dbcAMP as described above until the desired maturation stage for a given experiment (typically 5-7 days for transplantation studies or 32 days for in vitro functional studies).
  • RNeasy kit Qiagen, Valencia, Calif.
  • RNA at day 25 of each condition was reverse transcribed (Quantitech, Qiagen) and amplified material was detected using commercially available Taqman gene expression assays (Applied Biosystems, Carlsbad, Calif.) with the data normalized to HPRT. Each data point represents 9 technical replicates from 3 independent biological samples.
  • Raw data of microarray studies are not yet available at GEO worldwideweb.ncbi.nlm.nih.gov/geo). Animal Surgery. Rodent and monkey procedures were performed following NIH guidelines, and were approved by the local Institutional Animal Care and Use Committee (IACUC), the Institutional Biosafety Committee (IBC) as well as the Embryonic Stem Cell Research Committee (ESCRO).
  • IACUC Institutional Animal Care and Use Committee
  • IBC Institutional Biosafety Committee
  • ESCRO Embryonic Stem Cell Research Committee
  • NOD-SCID IL2Rgc null mice (20-35 g in weight; Jackson Laboratory, Bar Harbor, Me.) were anesthetized with Ketamine (90 mg/kg; Akorn, Decatur, Ill.) and Xylazine (4 mg/kg Fort Dodge, Iowa).
  • 6-hydroxydopamine (10 ⁇ g (ranging in concentration from 0.1-20 ⁇ g) 6-OHDA (Sigma-Aldrich) was injected stereotactically into the striatum at the following coordinates (in millimeters): AP, 0.5 (from bregma; a skull suture used as reference for stereotactic surgery); ML, ⁇ 2.0; DV, ⁇ 3.0 (from dura a membrane covering the brain used for reference).
  • mice with successful lesions were selected for transplantation.
  • a total of 150 ⁇ 10 3 cells were injected in a volume of 1.5 ⁇ l into the striatum at the following coordinates (in mm): AP, 0.5; ML, ⁇ 1.8; DV, 3.2.
  • the mice were sacrificed 18 weeks post transplantation.
  • Rats were anesthetized with Ketamine (90 mg/kg) and xylazine (4 mg/kg) during surgical procedures. Unilateral, medial forebrain bundle lesions of the nigro-striatal pathway were established by stereotaxic injection of 6-OHDA (3.6 mg/ml in 0.2% ascorbic acid and 0.9% saline, Sigma) at two sites (Studer, et al. Nature Neurosci. 1:290-295 (1998), herein incorporated by reference). Rats were selected for transplantation if amphetamine-induced rotation exceeded 6 rotations/min by 6-8 weeks post injection.
  • Bilateral injections of cells (10 ul/injection; 125,000 cell/ul) were performed at three sites (1-posterior caudate, 2-pre-commissural putamen and overlying white matter) for a total volume of 30 ⁇ l per hemisphere.
  • An infusion pump attached to a stereotaxic micromanipulator was utilized to deliver the cells at a rate of 1 ⁇ l/min though a 50 ⁇ l Hamilton syringe with 28 G needle. After the injections were completed, the needle was left in place for an additional 2-5 minutes to allow the infusate to diffuse off the needle tip before slowly retracting the syringe.
  • the animals received analgesics (buprenex, 0.01 mg/kg IM, BID for 72 hours post surgery; meloxicam, 0.1 mg/kg SQ, SID for 72 hours post surgery) as well as an antibiotic (cephazolin, 25 mg/kg IM, BID) until 72-hours post-surgery.
  • analgesics buprenex, 0.01 mg/kg IM, BID for 72 hours post surgery; meloxicam, 0.1 mg/kg SQ, SID for 72 hours post surgery
  • antibiotic cephazolin, 25 mg/kg IM, BID
  • the animals received cyclosporine A (Neoral, Sandimmune) orally (30 mg/kg tapered to 15 mg/kg) once daily beginning 48-hrs prior to surgery until sacrifice, one month following transplantation.
  • Amphetamine-induced rotations (mice and rats) and the stepping test (rat) were carried out before transplantation and 4, 8, 12, 18 weeks after transplantation.
  • Rotation behavior in mice was recorded 10 min after i.p. injection of d-amphetamine (10 mg/kg, Sigma) and recorded for 30 minutes.
  • Rotation behavior in rats was recorded 40 min after i.p. injection of d-amphetamine (5 mg/kg) and automatically assessed by the TSE VideoMot2 system (Germany). The data were presented as the average number of rotations per minute.
  • the stepping test was modified from Blume, et al. Exp. Neurol. 219:208-211 (2009) and Crawley, et al.
  • mice and rats Animals (mice and rats) received overdoses of Pentobarbital intraperitoneally (50 mg/kg) to induce deep anesthesia and were perfused in 4% paraformaldehyde (PFA). Brains were extracted, post-fixed in 4% PFA then soaked in 30% sucrose solutions for 2-5 days. They were sectioned on a cryostat after embedding in O.C.T. compound (Sakura-Finetek, Torrance, Calif.).
  • ketamine (10 mg/kg, Intramuscular (IM)) and pentobarbital (25 mg/kg, intravenous (IV)) via cardiac perfusion with heparinized 0.9% saline followed by fresh cold 4% PFA fixative (pH7.4).
  • brains were removed from the skull and post-fixed in 4% PFA, free-floating, for 24-36 hrs. They were then rinsed and re-suspended in 10% sucrose on a slow shaker at 4° C., and allowed to “sink”. The process was then repeated in 20% sucrose followed by 30% sucrose.
  • Whole brains were cut coronally into 40 ⁇ m serial sections on a frozen sledge microtome and stored free-floating in cryopreservative medium at ⁇ 20° Celcius.
  • BSA bovine serum albumin
  • DA was measured directly in the medium using the same detection system but following aluminum extraction of dopamine and its metabolites using a commercially available kit as described previously (Studer, et al. Brain Res. Bull. 41:143-150 (1996), herein incorporated by reference).
  • Electrophysiological recordings Cultures were transferred to a recording chamber on an upright microscope equipped with a 40 ⁇ water-immersion objective (Eclipse E600FN; Nikon); cultures were perfused with saline containing in mM: 125 NaCl, 2.5 KCl, 25 NaHCO 3 , 1.25 NaH 2 PO 4 , 2 CaCl, 1 MgCl 2 , and 25 glucose (34° C.; saturated with 95% 0 2 -5% CO 2 ; pH 7.4; 298 mOsm/L). The saline flow rate was 2-3 ml/min running through an in-line heater (SH-27B with TC-324B controller; Warner Instruments).
  • Neurons were visualized by video microscopy with a cooled-CCD digital camera (CoolSNAP ES 2 , Photometrics, Roper Scientific, Arlington, Ariz.). Cells selected for electrophysiological recordings had neuron-like shapes with fine branching neurites. Somatic whole-cell patch-clamp recordings in current clamp configuration were performed with a MultiClamp 700B amplifier (Molecular Devices). Signals were filtered at 1-4 kHz and digitized at 5-20 kHz with a Digidata 1440A (Molecular Devices). Recording patch electrodes were fabricated from filamented borosilicate glass (Sutter Instruments) pulled on a Flaming-Brown puller (P-97, Sutter Instruments) and had resistances of 4-6 M ⁇ in the bath.
  • Electrodes were filled with internal solution containing in mM: 135 K-MeSO 4 , 5 KCl, 5 HEPES, 0.25 EGTA, 10 phosphocroeatine-di(tris), 2 ATP-Mg, and 0.5 GTP-Na (pH 7.3, osmolarity adjusted to 290-300 mOsm/L).
  • the amplifier bridge circuit was adjusted to compensate for electrode resistance and monitored. Electrode capacitance was compensated. When series resistance increased >20% during the recording, the data were discarded because increased resistance suggested a partial technical failure during recordings.
  • FIG. 1 The percentages of marker positive cells at the floor plate (day 11) FIG. 1 , midbrain dopamine neuron precursor (day 25), FIG. 2 and mature DA neuron stages (day 50 or later) FIGS. 3 and 11 , were determined in samples derived from 3 independent experiments each. Images for quantification were selected in a uniform random manner and each image was scored first for the number of DAPI-positive nuclei, followed by counting the number of cells expressing the marker of interest. Data are presented as mean ⁇ SEM. Quantification of human cells (identified with anti-hNA) and TH+ neurons within grafts was performed on every tenth section where a graft was identifiable.
  • hESC medium for maintenance (1 liter): 800 mL DMEM/F12, 200 mL of Knockout Serum Replacement, 5 mL of 200 mM L-Glutamine, 5 mL of Pen/Strep, 10 mL of 10 mM MEM minimum non-essential amino 15 acids solution, 55 ⁇ M of 13-mercaptoethanol, and bFGF (final concentration is 4 ng/mL).
  • KSR medium for hESC differentiation (1 liter): 820 mL of Knock out DMEM, 150 mL of Knock out Serum Replacement, 10 mL of 200 mM L-Glutamine, 10 mL of Pen/Strep, 10 mL of 10 mM MEM, and 55 ⁇ M of 13-mercaptoethanol.
  • DMEM Dulbecco's Modification of Eagles Medium
  • FBS primary mouse embryo fibroblast
  • FBS primary mouse embryo fibroblast
  • This example describes the discovery of small molecules and contact timing for providing directed differentiation of FOXA2+LMX1A+DA neurons of the present inventions.
  • DA-like neurons were used in transplantation studies that resulted in concerns on the further use of these cells for therapeutic applications.
  • procedures described in Perrier et al., 2004 and Fasano et al., 2010, including MS5 neural induction resulted in rosette cell formation and were used to make Day 11 precursors, see FIGS. 2, 16 and 17 for examples, that were further used to derive DA-like neurons.
  • These neurons resulted from a low percentage of the precursor cells in the resulting Day 11 cell populations.
  • FIG. 16A There were very small numbers of surviving TH+ neuron at 4.5 moths after transplantation ( ⁇ 50 TH+ cells/animal) in grafts from rosette derived DA neuron precursors FIG. 16A .
  • GFP marked cells GFP was driven by a ubiquitous promoter
  • hNA+ green
  • co-express TH red
  • Panels 16 D-E show that D-E, despite the very poor in vivo survival there was some (low and highly variable) improvement in a few behavioral assays such as amphetamine induced rotations (D), cylinder test and spontaneous rotations (E).
  • Feeder-free neural induction was carried out as previously described (Chambers et al., 2009) but further modified to yield floor plate cells (Fasano et al., 2010).
  • the modified Dual-SMAD inhibition method for differentiating pluripotent cells into floor plate cells the inventors' previously discovered that high concentrations of SHH were required for FP induction by day 11. For example, in some embodiments, Sonic C25II was added at 200 ng/ml.
  • a cell population containing pluripotent cells was chosen by the inventors for a starting population and plated at Day 0.
  • the inventors followed a cell population with regular feedings containing fresh LSB until Day 11 and discovered that some remaining cells were LMX1A+ but did not express FOXA2 ( FIG. 1 a,b ).
  • the inventors plated duplicate starting cell populations then tested for cell types (i.e.
  • Three primary exemplary culture conditions tested were 1) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11, 2) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11 while during this time cells were additionally contacted with fresh purmorphamine, SHH and FGF8 until Day 7 and 3) cells contacted with LDN/SB (LSB) on Day 0 then contacted with fresh LSB until Day 5, on Day 5 cells were contacted with fresh LDN without SB until Day 11 while during this time cells were additionally contacted with fresh purmorphamine, SHH and FGF8 until Day 7 while additionally contacted with fresh CHIR starting on Day 3 of culture until Day 11 with several variations of these primary conditions in order to determine optimal yield of cell types.
  • FIG. 1 d Systematic comparisons of the three culture conditions were performed using global temporal gene expression profiling. See exemplary FIG. 8 and Tables 1-6.
  • Hierarchical clustering of differentially expressed genes segregated the three treatment conditions by day 11 of differentiation FIG. 8 a ).
  • FOXA1, FOXA2 and several other SHH downstream targets including PTCH1 were amongst the most differentially regulated transcripts in LSB/S/F8/CHIR versus LSB treatment sets ( FIG. 1 e ).
  • LSB cultures by day 11 were enriched for dorsal forebrain precursor markers such as HES5, PAX6, LHX2, and EMX2.
  • Direct comparison of LSB/S/F8/CHIR versus LSB/S/F8 treatment confirmed selective enrichment for midbrain DA precursor markers in LSB/S/F8/CHIR group and suggested hypothalamic precursor identity in LSB/S/F8 treated cultures based on the differential expression of RAX1, SIX3, and SIX6 (see also POMC, OTP expression in FIG. 2 d ).
  • Exemplary lists of differentially expressed transcripts for day 11 are shown in Tables 1, 2 and day 25 in Tables 3-5 and gene ontology analysis FIG. 8 b (DAVID; http://david.abcc.ncifcrf.gov) confirmed enrichment for canonical WNT signaling upon CHIR treatment.
  • Raw data are not yet available at GEO worldwideweb.ncbi.nlm.nih.gov/geo/ accession#: [TBD]).
  • Comparative temporal analysis of gene expression for midbrain DA precursor markers FIG. 1 g
  • markers of anterior and ventral non-DA neuron fates FIG.
  • LSB dorsal forebrain
  • LSB/S/F8 ventral/hypothalamic
  • LSB/S/F8/CHIR midbrain DA precursor identity
  • DA neurons Differentiation of DA neurons. For further differentiation, precursor cells were maintained in a medium promoting neuronal maturation (BAGCT—see material and methods). The following types of comparisons were made between the populations of differentiated cells resulting from previous methods and methods of the present inventions: A) Immunocytochemical analysis at day 50 of differentiation for TH in combination with LMX1A, FOXA2 and NURR1, B) Quantification of TH+, FOXA2+, LMX1+, and NURR1+ cells out of total cells comparing rosette-derived versus floor plate-derived (LSB/S/F8/CHIR) cultures.
  • LSB/S/F8/CHIR treatment yielded TH+ cells that co-expressed LMX1A and FOXA2 and displayed strong induction of the nuclear receptor NURR1 (NR4A2) ( FIG. 2 a,b ).
  • Comparing gene expression in day 13 versus day 25 cultures confirmed robust induction of other postmitotic DA neuron markers ( FIG. 2 c ).
  • Characterizing DA neuron identity at day 25 in comparison to LSB and LSB/S/F8 treated cultures confirmed enrichment for known midbrain DA neuron transcripts and identified multiple novel candidate markers ( FIG. 2 d , Tables 3-5, FIG. 8 b ).
  • the transcript most highly enriched in LSB/S/F8/CHIR was TTF3, a gene not previously associated with midbrain DA neuron development, but highly expressed in the human substantia nigra ( FIG. 8 c ; Allen Brain Atlas: http://human.brain-map.org).
  • FIGS. 10 and 16 In vitro and in vivo properties of floor plate-derived DA neurons were compared to DA-like neurons obtained via a neural rosette intermediate ( FIGS. 10 and 16 ). Patterning of neural rosettes represents the currently most widely used strategy for deriving DA neurons from hPSCs. Both floor plate- and rosette-based protocols were efficient at generating TH+ neurons capable of long-term in vitro survival (day 50 of differentiation; FIG. 3 a ). However, the percentage of TH+ cells was significantly higher in floor plate-derived cultures ( FIG. 3 b ). While TH+ cells in both protocols displayed co-expression of NURR1, floor plate-derived DA neurons co-expressed FOXA2 and LMX1A ( FIG. 3 a,b ).
  • GABA and serotonin (5-HT)-positive neurons were observed ( FIG. 3 c ).
  • DA, and its metabolites DOPAC and HVA were present in cultures generated with either protocol, but DA levels were approximately 8 times higher in floor plate cultures ( FIG. 3 d,e ).
  • Midbrain DA neurons exhibited extensive fiber outgrowth and robust expression of mature neuronal markers including synapsin, dopamine transporter (DAT), and G-protein coupled, inwardly rectifying potassium channel (Kir3.2—also called GIRK2—expressed in substantia nigra pars compacta (SNpc) DA neurons) ( FIG. 3 f , FIG. 11 ).
  • SNpc DA neurons in vivo exhibit an electrophysiological phenotype that differentiates them from most other neurons in the brain. In particular, they spike spontaneously at a slow (1-3 Hz) rate. Moreover, this slow spiking is accompanied by a slow, sub-threshold oscillatory potential. After 2-3 weeks in vitro, these same physiological features are displayed by SNpc DA neurons cultured from early postnatal mice. The DA neurons differentiated from hESCs consistently (4/4) displayed this distinctive physiological phenotype ( FIG. 3 g - i ).
  • FIG. 3A DA release measurement by HPLC showed d65 old TH+ neurons are functional in vitro FIG. 3B .
  • Engraftment of novel DA neuronal cell population in rodents i.e. mice and rats containing damaged neurons.
  • hPSC-derived midbrain DA neurons that functionally engraft in vivo without the risk of neural overgrowth or inappropriate differentiation into non-midbrain neurons or develop teratomas.
  • the time of cell cycle exit marked by expression of NURR1
  • FIG. 2 Initial studies using day 25 cells in non-lesioned adult mice showed robust survival of hPSC-derived FOXA2+/TH+ neurons at 6 weeks after transplantation ( FIG. 12 ).
  • the overgrowth was likely due to the longer survival periods (4.5 months versus 6 weeks), lack of FACS purification prior to transplantation and choice of NOD-SCID IL2Rgc null host.
  • the number of proliferating Ki67+ cells was minimal in floor plate-derived grafts ( ⁇ 1% of total cells), while rosette-derived grafts retained pockets of proliferating neural precursors.
  • Neural overgrowth is thought to be caused by primitive anterior neuroectodermal cells within the graft (Elkabetz, et al. Genes Dev. 22:152-165 (2008); Aubry, et al. Proc. Natl. Acad. Sci. U.S.A. 105:16707-16712 (2008), herein incorporated by reference).
  • results in NOD-SCID IL2Rgc null mice described herein demonstrated robust long-term survival of FOXA2+/TH+ neurons, complete reversal of amphetamine-induced rotation behavior and lack of any signs of neural overgrowth. However, some of these outcomes could be attributable to the specific use of NOD-SCID IL2Rgc null mice.
  • floor plate-derived DA neuron cultures 250 ⁇ 10 3 cells were transplanted in adult 6-OHDA lesioned rats immunosuppressed pharmacologically using cyclosporine A. Five months after transplantation graft survival was robust ( FIG. 4 e - h ) with an average of more than 15,000 TH+ cells co-expressing FOXA2 ( FIG.
  • FIG. 4 g TH+/hNCAM+ fibers emanated from the graft core into the surrounding host striatum ( FIG. 4 f ).
  • TH+ cells expressed midbrain DA neuron markers PITX3 and NURR1 ( FIG. 4 h - j ).
  • Behavioral analyses showed complete rescue of amphetamine-induced rotational asymmetry, in contrast to sham-grafted animals that did not show improvements ( FIG. 4 k ). Grafted animals also showed improvements in the stepping test ( FIG. 4 l ) measuring forelimb akinesia and in the cylinder test ( FIG.
  • mice As in mice ( FIG. 13 ), serotonergic and GABAergic cells were rare ( ⁇ 1% of total cells) in rat cells, as were the mostly host-derived GFAP+ glial cells (7% of total cells; ( FIG. 14 ). While few serotonin+ neurons were detected in the graft, hNCAM-negative cells were observed that were likely host-derived serotonergic fibers ( FIG. 14 ).
  • the graft cores were composed of TH+ neurons co-expressing SC-121 ( FIG. 4 s ) and FOXA2 ( FIG. 4 t ).
  • SC-121 and GFP negative areas within the graft contained Iba1+ host microglia ( FIG. 15 ) indicating incomplete immunosuppression.
  • engraftment of novel DA neuronal cell population in primates i.e. adult MPTP (3 mg of MPTP-HCL (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; ranging in concentration from 0.5-5 mg MPTP-HCl) lesioned rhesus monkeys containing a severe >95% loss of endogenous midbrain DA neurons.
  • MPTP exposure caused observable changes and symptoms similar to Parkinson's disease in humans.
  • This example described the discovery that large populations of midbrain DA neurons developed with characteristics of a PD patient's neurons when a PD patient's cell line, i.e. PINK1 mutant PD-iPSC cell, obtained in a manner that did not result in the destruction of an embryo, were used as the cell population for obtaining FOXA2/LIM1XA/TH+ DA neurons of the present inventions.
  • PINK1 Q456X mutant PD-iPSC line was differentiated using the novel floor-plate based midbrain DA neuron protocol (method) of the present inventions which yielded midbrain differentiation profiles comparable to those obtained from the iPSC H9 line.
  • FIG. 20 A-C) Immunocytochemical analysis of PINK1 mutant PD-iPSC line at day 11 of differentiation (midbrain precursor stage) for FOXA2 (red), LMX1A (green) and DAPI (blue) (A), day 25 of differentiation (early postmitotic DA neuronal stage) for FOXA2 (red) and TH (green) (B) and for NURR1 (red) and TH (green) (C).
  • PINK1 mutant PD-iPSC showed PD like phenotype of protein aggregation following long-term differentiation and maturation in vitro.
  • the inventors discovered that PINK1 mutant PD-iPSC showed evidence of ⁇ -synuclein (major component of Lewy body on PD patience) expression in cytosol of TH+ DA neurons at day 55 of differentiation using the novel floor-plate based midbrain DA neuron induction protocol, ( FIG. 21 a - b ).
  • A, B Immunocytochemical analysis of PINK1 mutant PD-iPSC line at day 55 of differentiation for ⁇ -synuclein (LB509, red), TH (green) and merged image (A) and ⁇ -synuclein (red) and ubiquitin (green) (B). These ⁇ -synuclein positive cells also showed high expression of ubiquitin (classical Lewy body marker). In contrast, DA neurons derived from control iPS line showed expression of normal synaptic (as opposed to cytosolic) ⁇ -synuclein expression and very low levels of Ubiquitin ( FIG. 21 c - d ).
  • ⁇ -synuclein expression of aggregated form of ⁇ -synuclein.
  • dimerized insoluble forms of a-synulcein lead to aggregation in Lewy bodies.
  • the dimerized form of ⁇ -synuclein shows phospholylation of Serine 129 on ⁇ -synuclein.
  • PINK1 mutant PD-iPSC derived cells showed strong expression for Ser129 phosphorylated ⁇ -synuclein in contrast to control-iPSC derived cells that showed very low levels of expression ( FIG. 22 ).
  • PINK1 mutant PD-iPSC derived cells showed strong expression for Ser129 phosphorylated ⁇ -synuclein in contrast to control-iPSC derived cells that showed very low levels of expression.
  • Exemplary DA neurons derived from PINK1 mutant PD-iPSC are more vulnerable to toxic stimulation.
  • PD-iPSC derived TH+ DA neurons derived via floor-plate based protocol were more vulnerable to toxin challenge (valinomycin: mitochondria ionophore, 5 uM (ranging in concentration from 1-10 uM), 48 hr) than control-iPSC derived cells.
  • valinomycin mitochondria ionophore, 5 uM (ranging in concentration from 1-10 uM), 48 hr) than control-iPSC derived cells.
  • TH+ neurons derived via the classic MS5 based protocol did not show differential vulnerability between PD- versus control-derived cells. ( FIG. 24 ).
  • A-F Representative TH immunocytochemistry at day 60 of differentiation: Normal condition (no toxin treatment) for both PD- and control-iPSC derived cells obtained via floor-plate based protocol (A, PD-iPSC derived cells shown), nearly complete degeneration of TH+ DA neurons in PD-iPSC following toxin treatment (B), partially degenerated TH+ DA neurons from control-iPSC(C). Entire cell viability assay with alamar-blue after 48 hrs of valinomycin treatment also showed differential cell survival in a specific dose range for toxin challenge (5 and 10 uM) when comparing PD-iPSC and control iPSC ( FIG. 25 ).
  • K-N low power images of immunocytochemistry for Tuj1 (red) and TH (green) by MS5 based protocol at day 60 of differentiation: PD-iPSC of normal (K), versus toxin challenge (L) conditions and control iPSC of normal (M), versus toxin challenge (N) conditions.
  • Exemplary quantification of cell viability-dose response assay for toxin challenge Cell viability assay with alamar-blue after 48 hrs of valinomycin treatment showed differential cell survival in a specific dose range for toxin challenge (5 and 10 uM) when comparing PD-iPSC and control iPSC (day 60 of floor-plate based differentiation). Note: this assay tests for overall cell death while the most dramatic effects were observed specifically in DA neurons (see FIG. 14 ). Therefore, alamar blue based quantification will likely underestimate the extent of the differential effect observed on DA neuron lineages.
  • Exemplary conditions were established for the in vivo recording of human pluripotent stem cell derived DA neurons in acute slice preparations; see exemplary results shown in FIG. 26 .
  • Electrophysiological measurements are contemplated for use in acute slice preparations, i.e. from biopsies of engrafted areas.
  • A9- versus A10 type graft-derived DA neurons will be identified in vivo based on testing for the autonomouse pacemaking activity that is specific to A9-type dopamine neurons that are most affected in PD. In other words, A10 type neurons do not have pacemaking activity
  • FIG. 26 Conditions were established for the in vivo recording of human pluripotent stem cell derived DA neurons in acute slice preparations, see, FIG. 26 .
  • grafted human DA neurons derived from pluripotent stem cells were measured for and discovered to have electrophysiological features typical of those seen in mouse substantia nigra pars compacta (SNpc), FIG. 26A where the top view shows reconstruction of a pacemaking neuron in the graft region.
  • Bottom shows an exemplary photomicrograph of a brain slice taken from the rat into which the hES-derived neurons were injected 9 months prior; the graft is outlined; a higher magnification image is shown inset at the bottom.
  • the slice was processed for tyrosine hydroxylase which shows up as white, FIG.
  • FIG. 26B shows an exemplary cell-attached patch recording from a putative DA neuron in the graft; Bottom shows an exemplary whole cell recording from the same cell. Recordings were made in the presence of glutamate and GABA receptor antagonists (50 ⁇ M AP5, 10 ⁇ M CNQX and 10 ⁇ M GABAzine) to eliminate synaptic input. These recordings demonstrated that the PS-derived neurons were autonomous pacemakers with normal intrasomatic voltage trajectories. Another neuron recorded in a graft sample had similar properties, FIG. 26C . For comparison, cell-attached and whole cell recordings from a dopaminergic neuron in SNpc of an adult mouse are shown.
  • GABA receptor antagonists 50 ⁇ M AP5, 10 ⁇ M CNQX and 10 ⁇ M GABAzine
  • CTx cortex
  • STr striatum
  • SNpc substantia nigra pars compacta
  • DA dopaminergic
  • Exemplary methods for identifying cell surface markers for use in methods of the present inventions were identified with these methods.
  • FIG. 27 a An unbiased gene expression screen in genetic reporter lines ( FIG. 27 a ) that found several candidate markers, including a marker, termed DCSM1, that is selectively expressed in midbrain DA neurons and appears to specially marker A9-type DA neurons ( FIG. 27 b ).
  • a second strategy is the use of a CD cell surface marker screen in hESC derived DA neurons testing 242 commercially available antibodies in 96 well format ( FIG. 27 c,d ). The results of such a screen ( FIG. 27 e ) led to the identification of at least 5 validated markers enriched in midbrain DA neurons including CD142, a marker that selectively marks Nurr1+ DA neuron stage ( FIG. 27 f ).
  • CD142 typically marked approximately 30% of the total cell population at day 25 of differentiation ( FIG. 28 a ).
  • Selectivity of CD142 for a Nurr1+ DA neuron stage was confirmed in multiple independent hESC and hiPSC lines ( FIG. 28 b ).
  • enrichment of CD142 positive cells resulted in selective depletion of undesired neuron subtypes such as GABA and Serotonergic neurons.
  • FIG. 28 c - f in vivo studies confirmed the ability of a CD142 positive cell population to give rise to high purity DA neuron grafts that overcame problems of contaminating GABA and Serotonergic neurons.
  • CD142 based selection of precursor cells is contemplated to further reduce the risk of introducing serotonergic neurons, a contaminating cell type that was implicated in failed human fetal tissue grafting as the potential source of the undesirable fetal tissue graft-induced dyskinesias.
  • This example describes methods for transformation of cells with human PST genes for increase PSA cell surface expression. This example also shows exemplary methods of using cells having increased PSA cell surface expression.
  • this example shows engineered PST genes into hESCs for increasing PSA expression on DA neurons.
  • a gene encoding the human polysialyl-transferase (hPST) was introduced into a hESC line (WA01) using a lentiviral vector (pLenty, Invitrogen). Twenty selected clones were expanded and analyzed for PST expression. PST-expressing hESC clones were differentiated to ensure that PST was not silenced in DA neurons. Quantification of PSA-NCAM at different stages of differentiation (day 0, 11, 25, and 50) was done using FACS analysis and immunofluorescence (Operetta). Positive clones were subjected to the suite of DA neuron QC parameters outlined in Table 7.
  • At least 3 clones that retain high, uniform levels of PSA-NCAM during differentiation and perform well in the QC parameters will advance to assessment of the neurite outgrowth in PST-overexpressing hESC-derived DA neurons
  • Selected control and PST-overexpressing hESC clones were differentiated into DA neurons using the standard protocol described herein, followed by cell fixation and analysis at days 25 and 50.
  • the number and length of TH-positive fibers in such cultures were quantified with the Operetta High Content Microscope.
  • PST-overexpressing and control hESC clones that advance from in vitro studies above, were differentiated again into DA neurons and transplanted into a rat model of PD. Short-term grafts (4-6 weeks) to determine survival, PSA-NCAM expression and neurite outgrowth were done. For each clone that passed short-term in vivo parameters were subjected to long-term grafting studies. For those studies animals received half or a quarter of the standard (200 ⁇ 10 3 ) dose of cells. These studies were to address whether increased PSA leads to increased long-term survival after transplantation (5 months), and whether smaller DA neuron numbers are capable of matching or outperforming the functional capacity of non-PST grafts transplanted at standard cell doses (not FIG. 27 ).
  • the following example shows enhancement of polysialic acid expression that improved the function of ES-derived dopamine neuron grafts in Parkinsonian mice.
  • ES cells expressing GFP under control of Nurr1 promoter were stably transduced with a lentiviral vector ubiquitously expressing polysialyltransferase (PST).
  • PST polysialyltransferase
  • Transduced cells showed a dramatic increase in PST mRNA as compared to controls ( FIG. 30A ).
  • Expression of PST was observed to be sufficient for PSA synthesis on NCAM. Accordingly, PSA-NCAM expression was greatly increased in PST-modified cells at day 14 of DA neuron differentiation ( FIG. 30B-E ). Both the endogenous and induced cell surface PSA on ES-derived DA neurons could be removed ( FIG.
  • PSA enhancement provided a significant augmentation of the ability of grafted DA neurons to innervate host striatum and attenuate PD functional deficits. Therefore clinical translation is contemplated comprising DA neurons of the present inventions for providing cells prior to transplantation.
  • the cells will be genetically manipulated for expressing PSA.
  • PST may be delivered directly to the cells via exposure to the purified enzyme and substrate, in vitro, prior to transplantation.
  • PSA strategy for human translation in PD grafting is contemplated to minimize the need for multiple injections and thereby reduce the surgical risks resulting from these multiple injections.
  • this technology is contemplated for use on other cell types and species, for example, augmenting the migration of grafted Schwann cells in creating a bridge (for example, cell-cell communication) for re-growth of axons at the site of spinal cord injury.
  • a bridge for example, cell-cell communication
  • mice Six-week old 129S3/SvImJ mice (Jackson Laboratory) were kept under controlled temperature with food and water available ad libitum. Experimental procedures were performed according to NIH and institutional animal use guidelines and approved by the local Institutional Animal Care and Use Committee (IACUC) and the Institutional Biosafety Committee (IBC).
  • IACUC Institutional Animal Care and Use Committee
  • IBC Institutional Biosafety Committee
  • 6OHDA injection and amphetamine-induced test Animals were anesthetized with sodium pentobarbital (10 mg/kg) and injected in the right striatum with 2 ⁇ l of 6OHDA (4 ⁇ g/ ⁇ l in saline, 0.5% ascorbic acid). The injections were performed with a Hamilton syringe at coordinates: 0.5 mm posterior, 1.8 mm lateral relative to bregma and 2.5 mm ventral to brain surface. Before the surgery animals received a single i.p. injection of desipramine (25 mg/Kg, Sigma). Two weeks after surgery animals were scored in the amphetamine-induced rotation test.
  • a Nurr1::GFP BAC transgenic BAC mouse ES reporter cell line i.e., GFP expression is driven by Nurr1 promoter
  • a lentivirus pLenti, Invitrogen
  • ES cells were propagated on mitomycin C-treated MEFs (StemCell Technologies) in DMEM (Invitrogen), 10% FBS (HyClone) supplemented with 1,400 units/ml LIF (ESGRO; Invitrogen), 2 mM L-glutamine, 1 mM ⁇ -mercaptoethanol, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin (Invitrogen).
  • DA differentiation was induced according to Barberi et al., Nat Biotechnol 21, 1200-1207 (2003), with modifications. Briefly, cells were differentiated on MS5 feeder cells in gelatin-coated dishes (10,000 cells/10 cm dish) and cultured for four days on serum replacement media (SRM).
  • SRM serum replacement media
  • Sonic hedgehog SHH, 200 ng/ml
  • FGF8 100 ng/ml
  • the media was changed to N2 supplemented with SHH, FGF8 and bFGF (10 ng/ml).
  • terminal differentiation was induced by withdrawal of SHH, FGF8 and bFGF and the addition of ascorbic acid (AA, 200 ⁇ M) and BDNF (20 ng/ml).
  • Nurr1::GFP sorted cells were analyzed for viability and resuspended in N2 with BDN and AA to a final concentration of 55,000 cells/ ⁇ l.
  • One ⁇ l was injected into the lesioned mouse striatum with a 50 ⁇ m tipped fine glass capillary at coordinates: 0.3 mm posterior, 1.5 mm lateral from bregma and 2.2 mm ventral to the brain surface.
  • An aliquot of the cell suspension was re-plated in matrigel-coated 6 mm dishes for further characterization.
  • cells were fixed with paraformaldehyde for 10 min at 40 C, washed twice with PBS, blocked with 5% BSA (0.1% Triton X-100 in PBS) and incubated with primary antibodies for 2 hrs at room temperature: rabbit anti-GFP (1:1000, Invitrogen), mouse IgM anti-PSA (1:2000, 5A5), mouse anti-NeuN (1:800, Chemicon), mouse anti-TH (1:1000, Sigma), goat anti-FoxA2 (1:800, Santa Cruz), goat anti-Engrailed (1:800, Santa Cruz). Cells were then incubated with Cy-conjugated secondary antibodies (1:1000, Jackson).
  • EndoN treatment To remove PSA from NCAM, the night before harvesting, cells were treated with 20 units of endoN, a phage enzyme that specifically removes PSA 7-9. Cells were then harvested and injected as described before but were resuspended in N2 with BDNF and AA and 5 units of endoN. We previously assessed that the injection of the same amount of endoN alone into lesioned mice did not improve animal behavior.
  • PST mRNA and PSA-NCAM analysis in vitro were treated with WB buffer (PBS with 1% NP40, 150 mM NaCl, 1 mM EDTA, and 1 ⁇ protease/phosphatase inhibitors added immediately before extraction, at pH of 7.4) and sonicated twice for 5 sec, centrifuged and resuspended in Laemli buffer (LB). Aliquots without LB were saved for protein determination. Equal amounts of protein were loaded into 6% sodium dodecyl sulfate-polyacrylamide gel electrophoresis gel (BioRad). Proteins were transferred by electrophoresis onto polyvinylidene membranes (Millipore).
  • WB buffer PBS with 1% NP40, 150 mM NaCl, 1 mM EDTA, and 1 ⁇ protease/phosphatase inhibitors added immediately before extraction, at pH of 7.4
  • LB Laemli buffer
  • the membranes were blocked for 1-6 hr in 0.1% Triton X-100 TBS (TBS-T) with 5% non-fat dry milk and incubated overnight with anti-NCAM antibody (1:10,000, Santa Cruz) in TBS-T with 5% milk. Blots were then incubated with peroxidase-conjugated secondary antibody (1:10,000, Jackson) and detected with ECL detection method (Amersham Pharmacia Biotech). Protein levels were quantified using ImageJ software.
  • PSA-NCAM FACS analysis cells were harvested with accutase treatment for 45 min, washed once and incubated with mouse IgM anti-PSA (1:250, 5A5) for 25 min on ice, washed once with N2 media and incubated with Cy3-conjugated anti-mouse-IgM (1:250, Jackson) for another 25 min on ice. Cells were washed once with N2 and resuspended with 0.1% BSA with 7AAD and analyzed in a FACS Calibur cell sorter. As control, no primary antibody was added.
  • Immunohistological and stereological procedures Free floating coronal sections were blocked in 0.1% Triton X-100, 5% donkey serum in PBS for 30 min at room temperature and incubated 48 hrs at 4° C. with different antibodies: rabbit anti-GFP (1:300), chicken anti-GFP (1:200, Chemicon), mouse anti-TH (1:200), mouse IgM anti-PSA (1:1000), mouse anti-NeuN (1:400), goat anti-FoxA2 (1:300), rabbit anti-Girk2 (1:300, Alomone Labs), mouse anti-synapsin (1:200, BD Transduction Laboratories).
  • the number GFP+ and TH+ cells was counted in one-in-three sections encompassing the whole brain under a 40 ⁇ objective, and the total number of cells/graft estimated. Double-labeled cells were analyzed in single optical planes through the entire z-axis.
  • GFP+ cells were analyzed for each marker.
  • process outgrowth analysis confocal z-scans were performed at 0.8 ⁇ m intervals through the entire z-axis (20-40 ⁇ m) with a pinhole of 1 ⁇ m under a 40 ⁇ objective. Sections were scanned from the injection site laterally until no processes were observed. 3-D projections encompassing the whole scanned area were sequentially matched.
  • GFP and TH intensity analysis the entire scanned area was divided into five successive 100 ⁇ m zones away from the transplant and the intensities were measured using ImageJ software. Data were normalized to the intensity in the zone nearest the graft (zone I) to control for any potential differences in graft size.
  • the following example shows enzymatic engineering of PSA on hESC-derived DA neurons using the purified bacterial polysialyltransferase, PSTnm, to enhance transplant efficacy.
  • DA neurons externally treated with PSTnm is contemplated for use in the producing cells for engraftment.
  • Both mammalian PST and PSTnm produced chemically identical chains of PSA.
  • Increased PSA on hESC-derived DA neurons should persist for several weeks, sufficient for DA fibers to exit graft core. Because PSTnm is removed prior to grafting, immunogenicity to this enzyme contaminating the grafted cells should not be factor.
  • PSTnm was produced from an engineered fragment with enhanced solubility and activity characteristics (Willis et al., Characterization of the alpha-2,8-polysialyltransferase from Neisseria meningitidis with synthetic acceptors, and the development of a self-priming polysialyltransferase fusion enzyme. Glycobiology 18, 177-186 (2008)). Cultures of hESC were induced to differentiate into DA neurons before PSTnm exposure, exposure to substrate or both. Cultures were examined at different time-points of exposure (10 min to 6 hrs) by quantitative immunofluorescence (Operetta) and western blotting to determine the speed and levels of polysialylation.
  • Day 25 differentiated hESC-derived DA neurons will be incubated with the optimum concentrations of PSTnm and substrate using the conditions described herein.
  • PSA+ mDA neurons will be transplanted in short- and long-term assays as described herein and in FIG. 29 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Neurology (AREA)
  • Organic Chemistry (AREA)
  • Neurosurgery (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Psychology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Epidemiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Macromolecular Compounds Obtained By Forming Nitrogen-Containing Linkages In General (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Materials For Medical Uses (AREA)
US14/356,042 2011-11-04 2012-11-02 Midbrain dopamine (DA) neurons for engraftment Active US10280398B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/356,042 US10280398B2 (en) 2011-11-04 2012-11-02 Midbrain dopamine (DA) neurons for engraftment

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161555828P 2011-11-04 2011-11-04
PCT/US2012/063339 WO2013067362A1 (fr) 2011-11-04 2012-11-02 Neurones dopaminergiques (da) du mésencéphale pour greffe
US14/356,042 US10280398B2 (en) 2011-11-04 2012-11-02 Midbrain dopamine (DA) neurons for engraftment

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/063339 A-371-Of-International WO2013067362A1 (fr) 2011-11-04 2012-11-02 Neurones dopaminergiques (da) du mésencéphale pour greffe

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/353,546 Continuation US10711243B2 (en) 2011-11-04 2019-03-14 Midbrain dopamine (DA) neurons for engraftment

Publications (2)

Publication Number Publication Date
US20150010514A1 US20150010514A1 (en) 2015-01-08
US10280398B2 true US10280398B2 (en) 2019-05-07

Family

ID=48192829

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/356,042 Active US10280398B2 (en) 2011-11-04 2012-11-02 Midbrain dopamine (DA) neurons for engraftment
US16/353,546 Active US10711243B2 (en) 2011-11-04 2019-03-14 Midbrain dopamine (DA) neurons for engraftment
US16/892,000 Active 2034-06-24 US11970712B2 (en) 2011-11-04 2020-06-03 Midbrain dopamine (DA) neurons for engraftment

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/353,546 Active US10711243B2 (en) 2011-11-04 2019-03-14 Midbrain dopamine (DA) neurons for engraftment
US16/892,000 Active 2034-06-24 US11970712B2 (en) 2011-11-04 2020-06-03 Midbrain dopamine (DA) neurons for engraftment

Country Status (16)

Country Link
US (3) US10280398B2 (fr)
EP (2) EP3693455A1 (fr)
JP (6) JP6461602B2 (fr)
KR (4) KR102581669B1 (fr)
CN (5) CN110684734A (fr)
AU (3) AU2012332269B2 (fr)
BR (2) BR122020000182B8 (fr)
CA (2) CA3177246A1 (fr)
DK (1) DK2773748T3 (fr)
EA (1) EA201490796A1 (fr)
ES (1) ES2779453T3 (fr)
HK (1) HK1201879A1 (fr)
IL (4) IL286793B (fr)
PL (1) PL2773748T3 (fr)
PT (1) PT2773748T (fr)
WO (1) WO2013067362A1 (fr)

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010096496A2 (fr) 2009-02-17 2010-08-26 Memorial Sloan-Kettering Cancer Center Procédés de conversion neurale de cellules souches embryonnaires humaines
CA2831609C (fr) 2011-03-30 2019-06-11 Cellular Dynamics International, Inc. Amorcage de cellules souches pluripotentes pour la differentiation neurale
WO2013067362A1 (fr) 2011-11-04 2013-05-10 Memorial Sloan-Kettering Cancer Center Neurones dopaminergiques (da) du mésencéphale pour greffe
EP3498824A1 (fr) 2013-04-26 2019-06-19 Memorial Sloan-Kettering Cancer Center Interneurones corticaux et autres cellules neuronales produits par la différentiation dirigée de cellules pluripotentes et multipotentes
EP3031908B1 (fr) 2013-08-06 2018-12-05 Takeda Pharmaceutical Company Limited Méthode de production de neurones dopaminergiques
WO2015069736A1 (fr) * 2013-11-08 2015-05-14 The Mclean Hospital Corporation Procédés de génération efficace d'interneurones gabaergiques à partir de cellules souches pluripotentes
ES2939807T3 (es) * 2014-03-21 2023-04-27 Fujifilm Cellular Dynamics Inc Producción de neuronas dopaminérgicas del mesencéfalo y métodos para su utilización
WO2015143622A1 (fr) * 2014-03-25 2015-10-01 云南中科灵长类生物医学重点实验室 Milieu pour l'établissement de cellules souches neuro-épithéliales, procédé et utilisation
WO2016028880A1 (fr) 2014-08-19 2016-02-25 Cellular Dynamics International, Inc. Réseaux neuronaux formés à partir de cellules issues de cellules souches pluripotentes
US9730975B2 (en) * 2014-11-25 2017-08-15 The Penn State Research Foundation Chemical reprogramming of human glial cells into neurons for brain and spinal cord repair
EP3061809B1 (fr) 2015-02-27 2019-01-30 Miltenyi Biotec GmbH Procédé de production d'une composition cellulaire de cellules progénitrices dopaminergiques mésencéphaliques
EP3281151B1 (fr) * 2015-04-10 2023-03-29 President and Fellows of Harvard College Procédés et dispositifs pour analyse d'imagerie de cellule vivante
US10081791B2 (en) 2015-05-07 2018-09-25 William J. Freed Modeling connections between dopaminergic neurons and the cerebral cortex
EP4070803A1 (fr) 2015-06-01 2022-10-12 Memorial Sloan-Kettering Cancer Center Procédés de différenciation in vitro de neurones dopaminergiques mésencéphaliques (mda)
CN108368486B (zh) 2015-10-07 2023-06-27 纪念斯隆-凯特琳癌症中心 鉴定神经肌肉接头活动的调节剂的体外方法
CN105567642B (zh) * 2016-02-01 2019-07-12 中国科学院生物物理研究所 一种着色性干皮病人多能干细胞的制备方法
KR20180105236A (ko) 2016-02-05 2018-09-27 메모리얼 슬로안-케터링 캔서 센터 줄기 세포-유래 외배엽 계통 전구체의 분화 방법
WO2017176779A1 (fr) * 2016-04-05 2017-10-12 The Regents Of The University Of California Hydrogels cellularisés et ses procédés d'utilisation
CN105861532A (zh) * 2016-04-08 2016-08-17 北京大学深圳医院 一种基于miRNA491调节DAT基因表达的方法
KR101870240B1 (ko) * 2016-04-12 2018-06-22 서울대학교산학협력단 지방줄기세포에서 신경줄기세포, 신경세포 및 가바성 신경세포로의 분화 유도 방법
CN109072198B (zh) * 2016-04-22 2022-08-26 国立大学法人京都大学 产多巴胺神经祖细胞的制备方法
AU2017313065B2 (en) * 2016-08-16 2021-06-24 Fujifilm Cellular Dynamics Inc. Methods for differentiating pluripotent cells
FR3058892B1 (fr) * 2016-11-23 2021-04-09 Univ Bordeaux Unite de tissu neural et utilisation d'une telle unite pour l'implantation dans le systeme nerveux d'un mammifere
EP3600360A4 (fr) * 2017-03-21 2021-04-21 Memorial Sloan-Kettering Cancer Center Astrocytes dérivés de cellules souches, leurs procédés de préparation et procédés d'utilisation
AU2018272068A1 (en) * 2017-05-25 2020-01-16 Icahn School Of Medicine At Mount Sinai Method and composition for generating basal forebrain cholinergic neurons (BFCNs)
CN107828728A (zh) * 2017-10-26 2018-03-23 中山大学 一种制备中脑多巴胺神经元的方法
KR102201417B1 (ko) * 2018-05-02 2021-01-11 (주) 에스바이오메딕스 도파민 신경세포의 분리방법 및 이를 이용하여 분리된 도파민 신경세포를 포함하는 파킨슨병 치료용 약제학적 조성물
KR102111030B1 (ko) * 2018-05-10 2020-05-14 한국과학기술원 Jab1을 포함하는 신경세포 분화 탐지용 바이오마커 또는 신경세포 분화 촉진용 조성물
JP2022511568A (ja) * 2018-12-11 2022-01-31 ユニバーシティー オブ ロチェスター 統合失調症及び他の神経精神障害の治療方法
CN112011510B (zh) * 2019-05-30 2023-01-03 中国科学院上海有机化学研究所 一种由人多能干细胞制备多巴胺神经元的方法
US20220254448A1 (en) 2019-07-25 2022-08-11 The Scripps Research Institute Methods of identifying dopaminergic neurons and progenitor cells
KR102183208B1 (ko) * 2019-07-25 2020-11-25 한국과학기술연구원 Ngn2(neurogenin-2)를 이용하여 척수 손상 모델에서 반응성 성상세포를 신경세포로 리프로그래밍하는 방법
US20220389378A1 (en) * 2019-11-22 2022-12-08 Novo Nordisk A/S Spin-aggregated neural microspheres and the application thereof
MX2022011831A (es) 2020-03-25 2023-01-04 Sana Biotechnology Inc Células neurales hipoinmunogénicas para el tratamiento de trastornos y afecciones neurológicas.
CN115768875A (zh) * 2020-04-02 2023-03-07 纪念斯隆-凯特琳癌症中心 产生中脑多巴胺神经元的方法、中脑神经元及其用途
WO2021216623A1 (fr) 2020-04-21 2021-10-28 Aspen Neuroscience, Inc. Édition génique de lrrk2 dans des cellules souches et procédé d'utilisation de cellules différenciées à partir de celles-ci
WO2021216622A1 (fr) 2020-04-21 2021-10-28 Aspen Neuroscience, Inc. Édition génique de gba1 dans des cellules souches et procédé d'utilisation de cellules différenciées à partir de celles-ci
CN114292814A (zh) * 2020-09-22 2022-04-08 上海跃赛生物科技有限公司 一种多能干细胞分化为中脑黒质多巴胺能神经细胞的方法
AU2022256048A1 (en) 2021-04-07 2023-10-05 FUJIFILM Cellular Dynamics, Inc. Dopaminergic precursor cells and methods of use
CN117500916A (zh) 2021-04-16 2024-02-02 博恩赛尔公司 制备、扩增和纯化中脑多巴胺能祖细胞的方法
CN117202914A (zh) * 2021-04-21 2023-12-08 上海跃赛生物科技有限公司 一种质控和富集人多巴胺能神经前体细胞的方法
CN113388580B (zh) * 2021-06-16 2022-02-18 中国医学科学院医学实验动物研究所 一种诱导脂肪干细胞分化为功能性多巴胺能神经元的方法及用途
EP4355864A1 (fr) * 2021-06-18 2024-04-24 Memorial Sloan Kettering Cancer Center Procédés de génération de lignées de crête neurale sacrée et leurs utilisations
EP4370164A1 (fr) * 2021-07-14 2024-05-22 Innopeutics Corporation Composition destinée à inhiber l'agrégation d'alpha-synucléine et procédé d'inhibition de l'agrégation d'alpha-synucléine
WO2023004370A1 (fr) 2021-07-21 2023-01-26 Aspen Neuroscience, Inc. Modulation à base d'aav de gba1 et compositions associées et leurs utilisations
WO2023004371A1 (fr) * 2021-07-21 2023-01-26 Aspen Neuroscience, Inc. Procédés de différenciation de cellules neuronales et de prédiction de la prise de greffe de celles-ci et compositions associées
US20230081881A1 (en) 2021-07-21 2023-03-16 Aspen Neuroscience, Inc. Transposon-based modulation of gba1 and related compositions and uses thereof
CN113481160B (zh) * 2021-08-20 2022-04-29 北京太东生物科技有限公司 视网膜色素上皮细胞诱导培养基及其应用
KR20240055838A (ko) 2021-09-10 2024-04-29 후지필름 셀룰러 다이내믹스, 인코포레이티드 유도된 만능 줄기 세포 유래된 세포의 조성물 및 이의 사용 방법
CN117089520A (zh) * 2021-10-28 2023-11-21 中国科学院动物研究所 一种中脑多巴胺细胞群、其制造方法以及用途
CA3235390A1 (fr) 2021-10-29 2023-05-04 Deepika Rajesh Neurones dopaminergiques comprenant des mutations et leurs procedes d'utilisation
WO2023104792A1 (fr) 2021-12-07 2023-06-15 Vib Vzw Amplificateurs de maturation neuronale
WO2023177919A1 (fr) * 2022-03-18 2023-09-21 Memorial Sloan-Kettering Cancer Center Procédés et compositions d'amélioration de la survie in vivo de neurones dopaminergiques du mésencéphale
US20230377685A1 (en) 2022-04-15 2023-11-23 Aspen Neuroscience, Inc. Methods of classifying the differentiation state of cells and related compositions of differentiated cells
CN115521903B (zh) * 2022-09-30 2024-05-17 中国科学院生态环境研究中心 体外诱导人多能干细胞分化为多巴胺能神经元的方法

Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5166065A (en) 1988-08-04 1992-11-24 Amrad Corporation Limited In vitro propagation of embryonic stem cells
US5340740A (en) 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
US5453357A (en) 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5523226A (en) 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
WO2003000868A1 (fr) 2001-06-21 2003-01-03 Geron Corporation Neurones dopaminergiques et cellules precuseurs competentes en proliferation destines au traitement de la maladie de parkinson
US20030036195A1 (en) 2001-04-20 2003-02-20 Lorenz Studer Generation of differentiated tissue from nuclear transfer embryonic stem cells and methods of use
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
US20040087478A1 (en) 2000-08-03 2004-05-06 Clemens Gillen Screening method
US6787356B1 (en) 1998-07-24 2004-09-07 The United States Of America As Represented By The Department Of Health And Human Services Cell expansion system for use in neural transplantation
US20040214324A1 (en) 2002-12-09 2004-10-28 Ole Isacson Dopaminergic neurons differentiated from embryonic cells for treating neurodegenerative diseases
US6833269B2 (en) 2000-05-17 2004-12-21 Geron Corporation Making neural cells for human therapy or drug screening from human embryonic stem cells
US6887706B2 (en) 2001-10-03 2005-05-03 Wisconsin Alumni Research Foundation Method of in vitro differentiation of transplantable neural precursor cells from primate embryonic stem cells
US20050260747A1 (en) 2000-03-14 2005-11-24 Reubinoff Benjamin E Embryonic stem cells and neural progenitor cells derived therefrom
US7005252B1 (en) 2000-03-09 2006-02-28 Wisconsin Alumni Research Foundation Serum free cultivation of primate embryonic stem cells
US7011828B2 (en) 2000-03-14 2006-03-14 Es Cell International Pte. Ltd. Implanting neural progenitor cells derived for human embryonic stem cells
EP1645286A1 (fr) 2003-04-18 2006-04-12 Kyowa Hakko Kogyo Co., Ltd. Medicament pour la regeneration des nerfs
US7112437B2 (en) 2000-06-20 2006-09-26 Es Cell International Pte Ltd. Methods of culturing embryonic stem cells and controlled differentiation
US7211434B2 (en) 2000-09-29 2007-05-01 Van Der Kooy Derek Primitive neural stem cells and method for differentiation of stem cells to neural cells
US7252995B2 (en) 2002-12-13 2007-08-07 Yu-Show Fu Method of generating neurons from stem cells and medium for culturing stem cells
US20070224650A1 (en) 2002-07-16 2007-09-27 Thomas Jessell Systems and methods for screening for modulators of neural differentiation
WO2007113505A2 (fr) 2006-03-30 2007-10-11 The University Court Of The University Of Edinburgh Milieu de culture contenant des inhibiteurs de kinase et utilisation de celui-ci
US7294510B2 (en) 2001-03-30 2007-11-13 Japan Science And Technology Corporation Process for producing nerve stem cells, motor neurons, and GABAergic neurons from embryonic stem cells
US7297539B2 (en) 2000-01-11 2007-11-20 Geron Corporation Medium for growing human embryonic stem cells
WO2008018190A1 (fr) 2006-08-09 2008-02-14 Biomaster, Inc. Cellules de la crête neurale dérivées de tissu adipeux
US7332336B2 (en) 2003-08-19 2008-02-19 Effector Cell Institute, Inc. Methods for inducing differentiation of pluripotent cells
US7368115B2 (en) 2002-07-31 2008-05-06 Stem Cell Therapeutics Inc. Method of enhancing neural stem cell proliferation, differentiation, and survival using pituitary adenylate cyclase activating polypeptide (PACAP)
US20090035285A1 (en) 2002-09-25 2009-02-05 Bresagen, Inc. Compositions and Methods for Enrichment of Neural Stem Cells Using Ceramide Analogs
WO2009099152A1 (fr) 2008-02-06 2009-08-13 Public University Corporation Yokohama City University Méthode de culture cellulaire et méthode de criblage
US20100009442A1 (en) 2006-09-22 2010-01-14 Yoshiki Sasai Stem Cell Culture Medium and Method
US20100093090A1 (en) 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
US20100099772A1 (en) 2006-11-20 2010-04-22 Bean Bruce P Methods, compositions, and kits for treating pain and pruritis
WO2010063848A1 (fr) 2008-12-05 2010-06-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédé et milieu pour la différenciation neurale de cellules pluripotentes
US7763463B2 (en) 2000-05-17 2010-07-27 Geron Corporation Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
WO2010096496A2 (fr) 2009-02-17 2010-08-26 Memorial Sloan-Kettering Cancer Center Procédés de conversion neurale de cellules souches embryonnaires humaines
WO2010141622A2 (fr) 2009-06-02 2010-12-09 Cornell University Procédé pour isoler, identifier et utiliser des cellules souches embryonnaires destinées à devenir des interneurones du cerveau antérieur
WO2011019092A1 (fr) 2009-08-12 2011-02-17 Kyoto University Procédé d'induction d'une différenciation de cellules souches pluripotentes en cellules précurseurs neurales
US7892830B2 (en) 2007-01-17 2011-02-22 Wisconsin Alumni Research Foundation Clonal culture of human pluripotent stem cells
WO2011108766A1 (fr) 2010-03-03 2011-09-09 Kyoto University Méthode pour diagnostiquer une maladie liée à un mauvais repliement des protéines au moyen de cellules nerveuses dérivées de cellules ips
CN102191221A (zh) 2010-03-17 2011-09-21 中国人民解放军第二军医大学东方肝胆外科医院 一种能无限自我更新的神经干细胞、其制备方法及其用途
WO2011159726A2 (fr) 2010-06-14 2011-12-22 The Scripps Research Institute Reprogrammation de cellules pour leur conférer un nouveau destin
US8153428B2 (en) 2000-01-11 2012-04-10 Geron Corporation Use of TGF beta superfamily antagonists and neurotrophins to make neurons from embryonic stem cells
US8323971B2 (en) 2006-11-09 2012-12-04 Cambridge Enterprise Limited Differentiation of pluripotent cells into primary germ layer progenitors
US20130108669A1 (en) 2010-04-16 2013-05-02 The Mclean Hospital Corporation Dopaminergic neurons differentiated from pluripotent stem cells and uses of thereof
US20130183674A1 (en) 2010-05-25 2013-07-18 Memorial Sloan-Kettering Cancer Center Method of nociceptor differentiation of human embryonic stem cells and uses thereof
US8551783B2 (en) 2009-11-06 2013-10-08 Industry-Academic Cooperation Foundation, Yonsei University Efficient and universal method for neural differentiation of human pluripotent stem cells
KR101331034B1 (ko) 2011-06-09 2013-11-19 연세대학교 산학협력단 미분화 줄기세포로부터 중뇌특이적 도파민 신경세포의 제조 방법
US20140199274A1 (en) 2011-05-20 2014-07-17 The Mclean Hospital Corporation Neuronal progenitor cells and uses
US20140248696A1 (en) 2013-03-01 2014-09-04 Wisconsin Alumni Research Foundation Methods of maintaining, expanding, and diffrentiating neuronal subtype specific progenitors
US20140314721A1 (en) 2013-02-15 2014-10-23 International Stem Cell Corporation Use of neural cells derived from human pluripotent stem cells for the treatment of neurodegenerative diseases
US8883502B2 (en) 2010-09-09 2014-11-11 The Regents Of The University Of California Expandable cell source of neuronal stem cell populations and methods for obtaining and using them
US20150010515A1 (en) 2012-01-11 2015-01-08 Max- Planck-Gesellschaft zur Förderung der Wissenschaften E.V. Mammalian neural plate border stem cells capable of forming neural tube and neural crest cell lineages including central and peripheral neurons
US20150010514A1 (en) 2011-11-04 2015-01-08 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (da) neurons for engraftment
US8932857B2 (en) 2010-06-15 2015-01-13 Kyoto University Method for selecting reduced differentiation resistance human induced pluripotent stem cells
US20150030570A1 (en) 2012-02-29 2015-01-29 Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Culture medium for preparing neural stem cells and use thereof
CA2923592A1 (fr) 2013-09-05 2015-03-12 Kyoto University Procede d'induction de cellules precurseurs neuronales produisant de la dopamine a partir de cellules souches pluripotentes
US20150086481A1 (en) 2011-07-28 2015-03-26 Memorial Sloan-Kettering Cancer Center Diagnosis and treatment of parkinson's disease
US20150087541A1 (en) 2012-04-24 2015-03-26 International Stem Cell Corporation Derivation of Neural Stem Cells and Dopaminergic Neurons from Human Pluripotent Stem Cells
WO2015054526A2 (fr) 2013-10-09 2015-04-16 The Regents Of The University Of California Procédés de production de cellules souches rétiniennes de mammifère et leurs utilisations
US20150265652A1 (en) 2014-03-21 2015-09-24 Cellular Dynamics International, Inc. Production of midbrain dopaminergic neurons and methods for the use thereof
WO2015143622A1 (fr) 2014-03-25 2015-10-01 云南中科灵长类生物医学重点实验室 Milieu pour l'établissement de cellules souches neuro-épithéliales, procédé et utilisation
US20150353888A1 (en) 2012-12-28 2015-12-10 Kyoto University Method for inducing astrocytes
US9249389B2 (en) 2008-06-17 2016-02-02 The Mclean Hospital Corporation Methods for isolating early neurons and neuroblasts
US20160108362A1 (en) 2011-12-14 2016-04-21 National Chung Hsing University Culture medium and method for inducing differentiation of pluripotent stem cells into neuroepithelial cells
WO2016063985A1 (fr) 2014-10-24 2016-04-28 大日本住友製薬株式会社 Procédé de production de tissu nerveux
US20160115448A1 (en) 2013-04-26 2016-04-28 Memorial Sloan-Kettering Cancer Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
US20160115444A1 (en) 2013-04-16 2016-04-28 Memorial Sloan-Kettering Cancer Center Age-modified cells and methods for making age-modified cells
US20160145582A1 (en) 2009-11-04 2016-05-26 Cellular Dynamics International, Inc. Episomal reprogramming with chemicals
WO2016162747A2 (fr) 2015-04-09 2016-10-13 Biolamina Ab Procédés et compositions de production de cellules dopaminergiques dérivées de cellules souches pour leur utilisation dans le traitement de maladies neurodégénératives
US20160326491A1 (en) 2013-11-21 2016-11-10 Memorial Sloan-Kettering Cancer Center Specification of functional cranial placode derivatives from human pluripotent stem cells

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA200401646B (en) * 2003-03-12 2004-06-07 Reliance Life Sciences Pvt Ltd Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells.
AU2005294173A1 (en) 2004-10-05 2006-04-20 Memorial Sloan Kettering Cancer Center Promotion of cell migration and axon regeneration in the CNS
CA2626728A1 (fr) * 2005-11-10 2007-05-24 Genervon Biopharmaceuticals L.L.C. Methode de differenciation mntf et de culture de cellules souches
WO2010042669A2 (fr) * 2008-10-07 2010-04-15 Ipierian, Inc. Compositions et procédés pour co-culture
CN102762718B (zh) * 2009-10-13 2015-06-17 干细胞技术公司 用于分化干细胞的重量克分子渗透压浓度控制
CN101935635A (zh) * 2010-07-30 2011-01-05 中国人民解放军第二军医大学 人骨髓多能干细胞的体外扩增和向多巴胺能神经元定向分化的方法

Patent Citations (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5166065A (en) 1988-08-04 1992-11-24 Amrad Corporation Limited In vitro propagation of embryonic stem cells
US5340740A (en) 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5453357A (en) 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5523226A (en) 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
US6787356B1 (en) 1998-07-24 2004-09-07 The United States Of America As Represented By The Department Of Health And Human Services Cell expansion system for use in neural transplantation
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US20040005704A1 (en) 1998-11-18 2004-01-08 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US20120322146A1 (en) 2000-01-11 2012-12-20 Carpenter Melissa K Neural cell populations from primate pluripotent stem cells
US8153428B2 (en) 2000-01-11 2012-04-10 Geron Corporation Use of TGF beta superfamily antagonists and neurotrophins to make neurons from embryonic stem cells
US8252586B2 (en) 2000-01-11 2012-08-28 Geron Corporation Neural cell populations from primate pluripotent stem cells
US7297539B2 (en) 2000-01-11 2007-11-20 Geron Corporation Medium for growing human embryonic stem cells
US7005252B1 (en) 2000-03-09 2006-02-28 Wisconsin Alumni Research Foundation Serum free cultivation of primate embryonic stem cells
US7011828B2 (en) 2000-03-14 2006-03-14 Es Cell International Pte. Ltd. Implanting neural progenitor cells derived for human embryonic stem cells
US20060078543A1 (en) 2000-03-14 2006-04-13 Reubinoff Benjamin E Neural progenitor cells derived from embryonic stem cells
US20050260747A1 (en) 2000-03-14 2005-11-24 Reubinoff Benjamin E Embryonic stem cells and neural progenitor cells derived therefrom
US7763463B2 (en) 2000-05-17 2010-07-27 Geron Corporation Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
US8252585B2 (en) 2000-05-17 2012-08-28 Geron Corporation Neural progenitor cell populations
US6833269B2 (en) 2000-05-17 2004-12-21 Geron Corporation Making neural cells for human therapy or drug screening from human embryonic stem cells
US7112437B2 (en) 2000-06-20 2006-09-26 Es Cell International Pte Ltd. Methods of culturing embryonic stem cells and controlled differentiation
US20040087478A1 (en) 2000-08-03 2004-05-06 Clemens Gillen Screening method
US7211434B2 (en) 2000-09-29 2007-05-01 Van Der Kooy Derek Primitive neural stem cells and method for differentiation of stem cells to neural cells
US7294510B2 (en) 2001-03-30 2007-11-13 Japan Science And Technology Corporation Process for producing nerve stem cells, motor neurons, and GABAergic neurons from embryonic stem cells
US20030036195A1 (en) 2001-04-20 2003-02-20 Lorenz Studer Generation of differentiated tissue from nuclear transfer embryonic stem cells and methods of use
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
WO2003000868A1 (fr) 2001-06-21 2003-01-03 Geron Corporation Neurones dopaminergiques et cellules precuseurs competentes en proliferation destines au traitement de la maladie de parkinson
US6887706B2 (en) 2001-10-03 2005-05-03 Wisconsin Alumni Research Foundation Method of in vitro differentiation of transplantable neural precursor cells from primate embryonic stem cells
US20070224650A1 (en) 2002-07-16 2007-09-27 Thomas Jessell Systems and methods for screening for modulators of neural differentiation
US7368115B2 (en) 2002-07-31 2008-05-06 Stem Cell Therapeutics Inc. Method of enhancing neural stem cell proliferation, differentiation, and survival using pituitary adenylate cyclase activating polypeptide (PACAP)
US20090035285A1 (en) 2002-09-25 2009-02-05 Bresagen, Inc. Compositions and Methods for Enrichment of Neural Stem Cells Using Ceramide Analogs
US20040214324A1 (en) 2002-12-09 2004-10-28 Ole Isacson Dopaminergic neurons differentiated from embryonic cells for treating neurodegenerative diseases
US8153422B2 (en) 2002-12-09 2012-04-10 The Mclean Hospital Corporation Dopaminergic neurons differentiated from embryonic cells for treating neurodegenerative diseases
US7252995B2 (en) 2002-12-13 2007-08-07 Yu-Show Fu Method of generating neurons from stem cells and medium for culturing stem cells
EP1645286A1 (fr) 2003-04-18 2006-04-12 Kyowa Hakko Kogyo Co., Ltd. Medicament pour la regeneration des nerfs
US7332336B2 (en) 2003-08-19 2008-02-19 Effector Cell Institute, Inc. Methods for inducing differentiation of pluripotent cells
WO2007113505A2 (fr) 2006-03-30 2007-10-11 The University Court Of The University Of Edinburgh Milieu de culture contenant des inhibiteurs de kinase et utilisation de celui-ci
WO2008018190A1 (fr) 2006-08-09 2008-02-14 Biomaster, Inc. Cellules de la crête neurale dérivées de tissu adipeux
US20100009442A1 (en) 2006-09-22 2010-01-14 Yoshiki Sasai Stem Cell Culture Medium and Method
US8323971B2 (en) 2006-11-09 2012-12-04 Cambridge Enterprise Limited Differentiation of pluripotent cells into primary germ layer progenitors
US20100099772A1 (en) 2006-11-20 2010-04-22 Bean Bruce P Methods, compositions, and kits for treating pain and pruritis
US7892830B2 (en) 2007-01-17 2011-02-22 Wisconsin Alumni Research Foundation Clonal culture of human pluripotent stem cells
WO2009099152A1 (fr) 2008-02-06 2009-08-13 Public University Corporation Yokohama City University Méthode de culture cellulaire et méthode de criblage
US20100093090A1 (en) 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
US9249389B2 (en) 2008-06-17 2016-02-02 The Mclean Hospital Corporation Methods for isolating early neurons and neuroblasts
US20110229441A1 (en) 2008-12-05 2011-09-22 Association Francaise Contre Les Myopathies Method and Medium for Neural Differentiation of Pluripotent Cells
WO2010063848A1 (fr) 2008-12-05 2010-06-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédé et milieu pour la différenciation neurale de cellules pluripotentes
US20120094381A1 (en) 2009-02-17 2012-04-19 Stuart Chambers Methods of neural conversion of human embryonic stem cells
US8642334B2 (en) 2009-02-17 2014-02-04 Memorial Sloan Kettering Cancer Center Methods of neural conversion of human embryonic stem cells
WO2010096496A2 (fr) 2009-02-17 2010-08-26 Memorial Sloan-Kettering Cancer Center Procédés de conversion neurale de cellules souches embryonnaires humaines
US9487751B2 (en) 2009-06-02 2016-11-08 Cornell University Method to isolate, identify, and use embryonic stem cells directed to forebrain interneuron fate
US20120148549A1 (en) 2009-06-02 2012-06-14 Sloan Kettering Institute For Cancer Research Method to isolate, identify, and use embryonic stem cells directed to forebrain interneuron fate
WO2010141622A2 (fr) 2009-06-02 2010-12-09 Cornell University Procédé pour isoler, identifier et utiliser des cellules souches embryonnaires destinées à devenir des interneurones du cerveau antérieur
US20120142093A1 (en) 2009-08-12 2012-06-07 Kyoto University Method for inducing differentiation of pluripotent stem cells into neural precursor cells
WO2011019092A1 (fr) 2009-08-12 2011-02-17 Kyoto University Procédé d'induction d'une différenciation de cellules souches pluripotentes en cellules précurseurs neurales
US20160145582A1 (en) 2009-11-04 2016-05-26 Cellular Dynamics International, Inc. Episomal reprogramming with chemicals
US8551783B2 (en) 2009-11-06 2013-10-08 Industry-Academic Cooperation Foundation, Yonsei University Efficient and universal method for neural differentiation of human pluripotent stem cells
WO2011108766A1 (fr) 2010-03-03 2011-09-09 Kyoto University Méthode pour diagnostiquer une maladie liée à un mauvais repliement des protéines au moyen de cellules nerveuses dérivées de cellules ips
CN102191221A (zh) 2010-03-17 2011-09-21 中国人民解放军第二军医大学东方肝胆外科医院 一种能无限自我更新的神经干细胞、其制备方法及其用途
US20130108669A1 (en) 2010-04-16 2013-05-02 The Mclean Hospital Corporation Dopaminergic neurons differentiated from pluripotent stem cells and uses of thereof
US9453198B2 (en) 2010-05-25 2016-09-27 Memorial Sloan Kettering Cancer Center Method of nociceptor differentiation of human embryonic stem cells and uses thereof
US20130183674A1 (en) 2010-05-25 2013-07-18 Memorial Sloan-Kettering Cancer Center Method of nociceptor differentiation of human embryonic stem cells and uses thereof
US20160201032A1 (en) 2010-05-25 2016-07-14 Memorial Sloan Kettering Cancer Center Method of nociceptor differentiation of human embryonic stem cells and uses thereof
WO2011159726A2 (fr) 2010-06-14 2011-12-22 The Scripps Research Institute Reprogrammation de cellules pour leur conférer un nouveau destin
US8932857B2 (en) 2010-06-15 2015-01-13 Kyoto University Method for selecting reduced differentiation resistance human induced pluripotent stem cells
US8883502B2 (en) 2010-09-09 2014-11-11 The Regents Of The University Of California Expandable cell source of neuronal stem cell populations and methods for obtaining and using them
US20140199274A1 (en) 2011-05-20 2014-07-17 The Mclean Hospital Corporation Neuronal progenitor cells and uses
KR101331034B1 (ko) 2011-06-09 2013-11-19 연세대학교 산학협력단 미분화 줄기세포로부터 중뇌특이적 도파민 신경세포의 제조 방법
US20150086481A1 (en) 2011-07-28 2015-03-26 Memorial Sloan-Kettering Cancer Center Diagnosis and treatment of parkinson's disease
US20150010514A1 (en) 2011-11-04 2015-01-08 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (da) neurons for engraftment
US20160108362A1 (en) 2011-12-14 2016-04-21 National Chung Hsing University Culture medium and method for inducing differentiation of pluripotent stem cells into neuroepithelial cells
US20150010515A1 (en) 2012-01-11 2015-01-08 Max- Planck-Gesellschaft zur Förderung der Wissenschaften E.V. Mammalian neural plate border stem cells capable of forming neural tube and neural crest cell lineages including central and peripheral neurons
US20150030570A1 (en) 2012-02-29 2015-01-29 Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Culture medium for preparing neural stem cells and use thereof
US20150087541A1 (en) 2012-04-24 2015-03-26 International Stem Cell Corporation Derivation of Neural Stem Cells and Dopaminergic Neurons from Human Pluripotent Stem Cells
US20150353888A1 (en) 2012-12-28 2015-12-10 Kyoto University Method for inducing astrocytes
US20140314721A1 (en) 2013-02-15 2014-10-23 International Stem Cell Corporation Use of neural cells derived from human pluripotent stem cells for the treatment of neurodegenerative diseases
US20140248696A1 (en) 2013-03-01 2014-09-04 Wisconsin Alumni Research Foundation Methods of maintaining, expanding, and diffrentiating neuronal subtype specific progenitors
US20160115444A1 (en) 2013-04-16 2016-04-28 Memorial Sloan-Kettering Cancer Center Age-modified cells and methods for making age-modified cells
US20160115448A1 (en) 2013-04-26 2016-04-28 Memorial Sloan-Kettering Cancer Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
CA2923592A1 (fr) 2013-09-05 2015-03-12 Kyoto University Procede d'induction de cellules precurseurs neuronales produisant de la dopamine a partir de cellules souches pluripotentes
WO2015054526A2 (fr) 2013-10-09 2015-04-16 The Regents Of The University Of California Procédés de production de cellules souches rétiniennes de mammifère et leurs utilisations
US20160326491A1 (en) 2013-11-21 2016-11-10 Memorial Sloan-Kettering Cancer Center Specification of functional cranial placode derivatives from human pluripotent stem cells
US20150265652A1 (en) 2014-03-21 2015-09-24 Cellular Dynamics International, Inc. Production of midbrain dopaminergic neurons and methods for the use thereof
WO2015143622A1 (fr) 2014-03-25 2015-10-01 云南中科灵长类生物医学重点实验室 Milieu pour l'établissement de cellules souches neuro-épithéliales, procédé et utilisation
WO2016063985A1 (fr) 2014-10-24 2016-04-28 大日本住友製薬株式会社 Procédé de production de tissu nerveux
WO2016162747A2 (fr) 2015-04-09 2016-10-13 Biolamina Ab Procédés et compositions de production de cellules dopaminergiques dérivées de cellules souches pour leur utilisation dans le traitement de maladies neurodégénératives

Non-Patent Citations (277)

* Cited by examiner, † Cited by third party
Title
"DAPI Nucleic Acid Stain," Molecular Probes, Invitrogen, Ltd. pp. 1-5 (2006).
Agarwal et al., "Efficient Differentiation of Functional Hepatocytes from Human Embryonic Stem Cells," Stem Cells, 26:1117-1127 (2008).
Andersson et al., "Identification of Intrinsic Determinants of Midbrain Dopamine Neurons," Cell 124:393-405 (2006).
Aoki, et al., "Sox1O Regulates the Development of Neural Crest-Derived Melanocytes in Xenopus," Developmental Biology, 259(1):19-33 (2003).
Bailey et al., "Sensory Organs: Making and Breaking the Pre-Placodal Region," Current Topics in Developmental Biology, 72:167-204 (2006).
Baker et al., "Establishing neuronal identity in vertebrate neurogenic placodes," Development, 127:3045-3056 (2000).
Baker et al., "Vertebrate cranial placodes I. Embryonic induction," Dev Biol., 232(1):1-61, pp. 1-30 (2001a).
Baker et al., "Vertebrate cranial placodes I. Embryonic induction," Dev Biol., 232(1):1-61, pp. 31-61(2001b).
Bansal et al., "Specific Inhibitor of FGF Receptor Signaling: FGF-2-Mediated Effects on Proliferation, Differentiation, and MAPK Activation Are Inhibited by PD 173074 in Oligodendrocyte-Lineage Cells," Journal of Neuroscience, Research, 74(4):486-493.
Barberi et al., "Derivation of engraftable skeletal myoblasts from human embryonic stem cells," Nat Med, 13(5):642-648 (2007).
Barberi et al., "Derivation of Multipotent Mesenchymal Precursors from Human Embryonic Stem Cells," PLoS Med, 2(6):e161 (2005).
Barberi et al., "Neural subtype specification of fertilization and nuclear transfer embryonic stem cells and application in parkinsonian mice," Nat Biotechnol. 10:1200-1207 (2003).
Bennett et al., "Regulation of Wnt Signaling During Adipogenesis," J Biol Chem., 277(34):30998-31004 (2002).
Bhattacharyya et al., "Hierarchy of regulatory events in sensory placode development," Curr Opin Genet Dev. 14(5):520-6 (2004).
Bouwmeester et al., "Cerberus is a head-inducing secreted factor expressed in the anterior endoderm of Spemann's organizer," Nature, 382:595-601 (1996).
Bradley et al., "Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines," Nature 309:255-256 (1984).
Brevini et al., "No shortcuts to pig embryonic stem cells," Theriogenology, 74:544-550 (2010).
Briscoe and Ericson, "The specification of neuronal identity by graded Sonic Hedgehog signalling," Semin Cell Dev Biol. 3:353-62 (1999).
Brivanlou and Darnell, "Signal Transduction and the Control of Gene Expression," Science, 295(5556):813-818 (2002).
Bystron et al., "The First Neurons of the Human Cerebral Cortex," Nat Neurosci., 9(7):880-886 (2006).
Cadigan and Liu, "Wnt Signaling: Complexity At the Surface," J Cell Sci., 119(Pt 3):395-402 (2006).
Cai et al., "BMP and TGF-β pathway mediators are critical upstream regulators of Wnt signaling during midbrain dopamine differentiation in human pluripotent stem cells," Developmental Biology, 376:62-73 (2013).
Callaerts et al., "PAX-6 in Development and Evolution," Annu. Rev. Neurosci. 20:483-532 (1997).
Chambers et al., "Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors," 2012, Nature Biotechnology 30(7):715-720.
Chambers et al., "Dual-SMAD Inhibition/WNT Activation-Based Methods to Induce Neural Crest and Derivatives from Human Pluripotent Stem Cells," Methods in Molecular Biology (2013) DOI 10.1007/7651_2013_59.
Chambers, S. M. et al., "Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling", Nature Biotechnology, vol. 27, No. 3, Mar. 1, 2009, pp. 275-280.
Charrier et al., "Dual origin of the floor plate in the avian embryo," Development 129:4785-4796 (2002).
Charron et al., "The Morphogen Sonic Hedgehog Is an Axonal Chemoattractant that Collaborates with Netrin-1 in Midline Axon Guidance," Cell 113:11-23 (2003).
Chen et al., "Immortalization and characterization of a nociceptive dorsal root ganglion sensory neuronal line," J Peripher Nerv Syst., 12(2):121-130 (2007).
Cooper, O. et al., "Differentiation of human ES and Parkinson's disease iPS cells into ventral midbrain dopaminergic neurons requires a high activity form of SHH, FGF8a and specific regionalization by retinoic acid", Molecular and Cellular Neurosciences, vol. 45, No. 3, Nov. 1, 2010, pp. 258-266.
Crawford et al., "The Notch Response Inhibitor DAPT Enhances Neuronal Differentiation in Embryonic Stem Cell-Derived Embryoid Bodies Independently of Sonic Hedgehog Signaling," 2007, Developmental Dynamics 236:886-892.
Cuny et al., "Structure-Activity Relationship Study of Bone Morphogenetic Protein (BMP) Signaling Inhibitors," Bioorg Med Chem Lett., 18(15):4388-4392 (2008).
D'Amour et al., "Efficient differentiation of human embryonic stem cells to definitive endoderm," Nat Biotechnol, 23(12):1534-1541 (2005).
Deng et al., "Specific and integrated roles of Lmx1a, Lmx1b and Phox2a in ventral midbrain development," Development 138:3399-3408 (2011).
Dennis et al., "DAVID: Database for Annotation, Visualization, and Integrated Discovery," Genome Biology 4:R60 (2003).
Dezawa et al., "Potential of Bone Marrow Stromal Cells in Applications for Neuro-Degenerative, Neuro-Traumatic and Muscle Degenerative Diseases," Current Neuropharmacology, 3:257-266 (2005).
Doble and Woodgett, "GSK-3: Tricks of the Trade for a Multi-Tasking Kinase," Journal of Cell Science, 116(7):1175-1186 (2003).
Doetschman et al. "Establishment of Hamster Blastocyst-Derived Embryonic Stem (ES) Cells," Dev Biol., 127:224-227 (1988).
Dorsky et al., "Control of Neural Crest Cell Fate by the Wnt Signalling Pathway," Nature, 396(6709):370-373 (1998).
Dovey et al., "Functional Gamma-Secretase Inhibitors Reduce Beta-Amyloid Peptide Levels in Brain," J Neurochem., 76(1):173-181 (2001).
Ebendal et al., "Bone Morphogenetic Proteins and Their Receptors: Potential Functions in the Brain," Journal of Neuroscience, Research, 51(2):139-146 (1998).
Eiraku et al., "Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals," Cell Stem Cell, 3:519-53 (2008).
El Maarouf et al., "Use of polysialic acid in repair of the central nervous system," PNAS 103(45):16989-16994 (2006).
Eldar-Finkelman, 2011, Frontiers in molecular Neuroscience, 4:32:1-18. (Year: 2011). *
Elkabetz et al., "Human ES Cell-Derived Neural Rosettes Reveal a Functionally Distinct Early Neural Stem Cell Stage," Genes Dev., 22(2):152-165 (2008).
Elkabetz et al., "Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage," Genes Dev., 22:152-165 Erratum (2008).
Erceg et al., "Human Embryonic Stem Cell Differentiation Toward Regional Specific Neural Precursors," Stem Cells, 27:78-87 (2009).
Ericson et al., "Two Critical Periods of Sonic Hedgehog Signaling Required for the Specification of Motor Neuron Identity," Cell, 87:661-673 (1996).
Evans et al., "Derivation and preliminary characterization of pluripotent cell lines from porcine and bovine blastocysts," Theriogenology, 33(1):125-128 (1990).
Evans et al., "Establishment in culture of pluripotential cells from mouse embryos," Nature, 292:154-156 (1981).
Extended European Search Report dated Mar. 10, 2015 issued in European Patent Application No. 12846715.6.
Fang et al., "Electrophysiological Differences Between Nociceptive and Non- Nociceptive Dorsal Root Ganglion Neurones in the Rat In Vivo," The Journal of Physiology, 565(3):927-943 (2005).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Fig. 1 (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Fig. 2 (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Fig. 3 (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Fig. 4 (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Fig. legends (2009).
Fasano et al., "Bmi-1 cooperates with Foxgl to maintain neural stem cell self-renewal in the fore brain," Genes Dev., 23:561-574 Suppl. Materials (2009).
Fasano et al., "shRNA knockdown of Bmi-1 reveals a critical role for p21-Rb pathway in NSC self-renewal during development," Cell Stem Cell, 1:87-99 (2007).
Fasano, C.A. et al., Efficient Derivation of Functional Floor Plate Tissue from Human Embryonic Stem Cells, Cell Stem Cell, Apr. 2, 2010, vol. 6, No. 4, pp. 336-347.
George et al., "Nociceptive Sensory Neurons Derive From Contralaterally Migrating, Fate-Restricted Neural Crest Cells," Nat Neurosci., 10(10):1287-1293 (2007).
Gerdes et al., "Production of a Mouse Monoclonal Antibody Reactive With a Human Nuclear Antigen Associated With Cell Proliferation," Int J Cancer, 31(1):13-20 (1983).
Gerrero et al., "Bm-3.0: A POU-Domain Protein Expressed in the Sensory, Immune, and Endocrine Systems That Functions on Elements Distinct From Known Octamer Motifs," PNAS USA, 90(22):10841-10845 (1993).
Giles et al., "Pluripotency of Cultured Rabbit Inner Cell Mass Cells Detected by Isozyme Analysis and Eye Pigmentation of Fetuses Following Injection into Blastocysts or Morulae," Mol. Reprod and Dev., 36:130-138 (1993).
Glinka et al., "Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction," Nature, 391:357-362 (1998).
Gordon, 2006, Journal of Biological Chemistry, 281:22429-22433. *
Grappe et al., "Structural basis of BMP signalling inhibition by the cystine knot protein Noggin," Nature, 420:636-642 (2002).
Graves et al., "Derivation and Characterization of Putative Pluripotential Embryonic Stem Cells from Preimplantation Rabbit Embryos," Mol. Reprod and Dev., 36:424-433 (1993).
Hemmati-Brivanlou et al., "Follistatin, an Antagonist of Activin, is Expressed in the Spemann Organizer and Displays Direct Neuralizing Activity," Cell, 77:283-295 (1994).
Hendzel et al., "Mitosis-Specific Phosphorylation of Histone H3 Initiates Primarily Within Pericentromeric Heterochromatin During G2 and Spreads in an Ordered Fashion Coincident With Mitotic Chromosome Condensation," Chromosoma., 106(6):348-360 (1997).
Hogan, "Bone Morphogenetic Proteins: Multifunctional Regulators of Vertebrate Development," Genes Dev., 10(13):1580-1594 (1996).
Huang et al., "Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists," Nucleic Acids Res., 37(1):1-13 (2009).
Huang et al., "Systematic and integrative analysis of large gene lists using DAVID Bioinformatics Resources," Nat Protoc., 4(1):44-57 (2009).
Hunter et al., "Retinoic acid stimulates neurite outgrowth in the amphibian spinal cord," PNAS USA, 88:3666-3670 (1991).
Iannaccone et al., "Pluripotent Embryonic Stem Cells from the Rat Are Capable of Producing Chimeras," Dev. Biol. 163:288-292 (1994).
International Search Report dated Mar. 29, 2013 in International Patent Application No. PCT/US2012/063339.
International Search Report dated Nov. 11, 2010 in International Patent Application No. PCT/US10/024487.
International search report dated Sep. 26, 2014 in International Application No. PCT/US2014/035760.
ISR PCT/US2011/037179 dated Feb. 24, 2012.
Ivanova et al., "Dissecting self-renewal in stem cells with RNA interference," Nature, 442:533-538 (2006).
Jeong et al., "A functional screen for sonic hedgehog regulatory elements across a 1 Mb interval identifies long-range ventral forebrain enhancers," Development, 133:7761-7772 (2005).
Jeong et al., "Distinct regulators of SHH transcription in the floor plate and notochord indicate separate origins for these tissues in the mouse node," Development, 130:3891-3902 (2003).
Jessell et al., "Polarity and patterning in the neural tube: the origin and function of the floor plate," Ciba Found Symp., 144:255-276, pp. 255-266 (1989a).
Jessell et al., "Polarity and patterning in the neural tube: the origin and function of the floor plate," CibaFound Symp., 144:255-276, pp. 267-276 (1989b).
Jessell et al., Polarity and patterning in the neural tube: the origin and function of the floor plate. Ciba Found Symp., (discussion) 144:276-280, 290-295 (1989).
Jessell, "Neuronal specification in the spinal cord: inductive signals and transcriptional codes," Nat Rev Genet., 1:20-29 (2000).
Jin et al., "MAPK/ERK and Wnt/β-Catenin pathways are synergistically involved in proliferation of Sca-1 positive hepatic progenitor cells", Biochemical and Biophysical Research Communications 409:803-807 (2011).
Joannides et al., "Automated Mechanical Passaging: A Novel and Efficient Method for Human Embryonic Stem Cell Expansion," Stem Cells, 24(2):230-235 (2006).
Joksimovic et al., "Wnt antagonism of SHH facilitates midbrain floor plate neurogenesis," Nat Neurosci., 12:125-131 (2009).
Joksimovic, M. et al., "Wnt antagonism of Shh facilitates midbrain floor plate neurogenesis", Developmental Biology, vol. 331, No. 2, Jul. 15, 2009, pp. 507-508.
Kakegawa et al, International Association for Dental Research, General Session and Exhibition, Jun. 28-Jul. 1, Abstract #0267, Brisbane Australia.
Katoh, Cancer Biology and Therapy, 2006, 5:1059-1064. *
Kawasaki et al., "Induction of Midbrain Dopaminergic Neurons from ES Cells by Stromal Cell-Derived Inducing Activity," Neuron, 28:31-40 (2000).
Kikuchi et al., "Multiplicity of the Interactions of Wnt Proteins and Their Receptors," Cell Signal, 19(4):659-671 (2007).
Kim et al., "Oct4-Induced Pluripotency in Adult Neural Stem Cells," Cell, 136:411-419 (2009).
Kim et al., "Robust Enhancement of Neural Differentiation From Human ES and iPS Cells Regardless of Their Innate Difference in Differentiation Propensity," Stem Cell Reviews and Reports, 6(2):270-281 (2010).
Kimura-Yoshida et al., "Crucial roles of Foxa2 in mouse anterior-posterior axis polarization via regulation of anterior visceral endoderm-specific genes," PNAS, 104:5919-59249 with Data Supplement Figs. Legends SFig. 5 and SFig. 6 (2006).
Kirkeby et al., "Predictive Markers Guide Differentiation to Improve Graft Outcome in Clinical Translation of hESC Based Therapy for Parkinson's Disease," Cell Stem Cell, available online Oct. 27, 2016 (2016), downloaded Feb. 2, 2017 from <http://dx.doi.org/10.1016/j.stem/2016.09.004>.
Kitao et al., "Neurogenesis of Subpopulations of Rat Lumbar Dorsal Root Ganglion Neurons Including Neurons Projecting to the Dorsal Column Nuclei," The Journal of Comparative Neurology, 371(2):249-257 (1996).
Kittappa et al, "The foxa2 Gene Controls the Birth and Spontaneous Degeneration of Dopamine Neurons in Old Age," PLoS Biol., 5(12):e325 (2007).
Kodama et al., "Neurogenic Potential of Progenitors Derived from Human Circulating CD14+ Monocytes," Immunol. Cell Biol., 84(2):209-217 (2006).
Kriks, S. et al., Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease, Nature, Nov. 6, 2011, vol. 480, No. 7378, pp. 547-551.
Laflamme et al., "Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts," Nat Biotechnol., 25(9):1015-1024 (2007).
Lee et al., "Directed Differentiation and Transplantation of Human Embryonic Stem Cell-Derived Motoneurons," Stem Cells, 25:1931-1939 (2007).
Lee et al., "Instructive Role of Wnt/B-Catenin in Sensory Fate Specification in Neural Crest Stem Cells," Science, 303(5660):1020-1023 (2004).
Lee et al., "Isolation and Directed Differentiation of Neural Crest Stem Cells Derived From Human Embryonic Stem Cells," Nat Biotechnol., 25(12):1468-1475 (2007).
Lee et al., "Modeling Pathogenesis and Treatment of Familial Dysautonomia using Patient-Specific iPSCs," Nature, 461(7262):402-406 (2009).
Lee et al., "The Expression and Posttranslational Modification of a Neuron-Specific B-Tubulin Isotype During Chick Embryo genesis," Cell Motility and the Cytoskeleton, 17(2):118-132 (1990).
Letinic et al., "Origin of GABAergic neurons in the human neocortex," Nature, 2002, vol. 417, No. 6889, pp. 645-649.
Li et al., "Directed differentiation of ventral spinal progenitors and motor neurons from human embryonic stem cells by small molecules," Stem Cells, 4:886-893 (2008).
Li et al., "Rapid induction and long-term self-renewal of primitive neural precursors from human embryonic stem cells by small molecule inhibitors," 2011, PNAS 108(20):8299-8304.
Li et al., "Specification of Motoneurons From Human Embryonic Stem Cells," Nat Biotechnol., 23(2):215-221 (2005).
Lin W. et al., "Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development", Genome & Development Control, Developmental Biology 333:386-396 (2009).
Lois et al., "Germline Transmission and Tissue-Specific Expression of Transgenes Delivered by Lentiviral Vectors," Science, 295:868-872 (2002).
Lupo et al., "Multiple roles of Activin/Nodal, bone morphogenetic protein, fibroblast growth factor and Wnt/β-catenin signaling in the anterior neural patterning of adherent human embryonic stem cell cultures," Open Biol., 3:120167 (2013) (13 pages).
Lyuksyutova et al., "Anterior-Posterior Guidance of Commissural Axons by Wnt-Frizzled Signaling," Science, 302:1984-1988 (2003).
Ma et al., "Neurogenin1 and Neurogenin2 Control Two Distinct Waves of Neurogenesis in Developing Dorsal Root Ganglia," Genes Dev., 13(13):1717-1728 (1999).
MacDonald (2009, Developmental Cell, 17:9-26). *
Marmigere and Ernfors, "Specification and Connectivity of Neuronal Subtypes in the Sensory Lineage," Nat Rev Neurosci., 8(2):114-127 (2007).
Maroof et al., "Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells", Cell Stem Cell, Epub. May 2, 2013, vol. 12, No. 5, pp. 559-572.
Maroof et al., "Prospective Isolation of Cortical Interneuron Precursors from Mouse Embryonic Stem Cells," J. Neurosci, 30(13):4667-4675 (2010).
Martin, "Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells," PNAS USA, 78(12):7634-7638 (1981).
Matise et al., "Gli2 is required for induction of floor plate and adjacent cells, but not most ventral neurons in the mouse central nervous system," Development, 125:2759-2770 (1998).
Mehler et al., "Bone Morphogenetic Proteins in the Nervous System," Trends Neurosci., 20(7):309-317 (1997).
Menendez et al., "Wnt signaling and a Smad pathway blockade direct the differentiation of human pluripotent stem cells to multi potent neural crest cells," PNAS, 2011, vol. 108, No. 48, pp. 19240-19245.
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Fig. 6 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Fig. 7 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Fig. 8 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Fig. 9 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Information Fig. 6 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Information Fig. 7 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Information Fig. 8 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Information Fig. 9 (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Information List (2003).
Mizuseki et al., "Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells," PNAS USA, 100:5828-5833 Supporting Materials and Methods (2003).
Mukhopadhyay et al., "Dickkopf1 is required for embryonic head induction and limb morphogenesis in the mouse," Dev Cell, 3:423-434 (2001).
Mullor et al., "Pathways and consequences: Hedgehog signaling in human disease," Trends Cell Biol 12:562-569 (2002).
Munoz et al., "Constraints to Progress in Embryonic Stem Cells from Domestic Species," Stem Cell Rev. and Rep., 5:6-9 (2009).
Munoz-Sanjuan et al., "Neural Induction, The Default Model and Embryonic Stem Cells," Nat Rev Neurosci., 3:271-280 (2002).
NIH (Stem Cells: Scientific Progress and Future Research Directions, Department of Health and Human Services, Chapter 1, pp. 1-4, Jun. 2001). *
Notarianni et al., "Maintenance and differentiation in culture of pluripotential embryonic cell lines from pig blastocysts," J. Reprod. Fert. Suppl., 41:51-56 (1990).
Oliveri, "Epigenetic dedifferentiation of somatic cells into pluripotency: cellular alchemy in the age of regenerative medicine?" Regen. Med., 2(5):795-816 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Data Supplement (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 1 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 2 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 3 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 4 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 5 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 6 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 7 (2007).
Ono et al., "Differences in neurogenic potential in floor plate cells along an anteroposterior location: midbrain dopaminergic neurons originate from mesencephalic floor plate cells," Development, 134:3213-3225 Supplemental Fig. 8 (2007).
Papapetrou et al., "Stoichiometric and Temporal Requirements of Oct4, Sox2, Klf4, and c-Myc Expression for Efficient Human Ipsc Induction and Differentiation," PNAS USA, 106(31):12759-12764 (2009).
Paris et al., "Equine embryos and embryonic stern cells: Defining reliable markers of pluripotency," Theriogenology, 74:516-524 (2010).
Patel et al., "Advances in Reprogramming Somatic Cells to Induced Pluripotent Stem Cells," Stem Cell Rev., 6(3):367-380 (2010).
Paterson et al., "Preclinical Studies of Fibroblast Growth Factor Receptor 3 As a Therapeutic Target in Multiple Myeloma," Br. J. Haematol., 124(5):595-603 (2004).
Patten et al., "Distinct modes of floor plate induction in the chick embryo," Development, 130:4809-4821 (2003).
Perrier et al., "Derivation of midbrain dopamine neurons from human embryonic stem cells," PNAS USA, 101:12543-12548 (2004).
Perrier et al., "Derivation of midbrain dopamine neurons from human embryonic stem cells," PNAS USA, 101:12543-12548 Supporting Materials, Methods, Supplemental Data, and Supporting Fig. 6 (2004).
Placantonakis et al., "BAC Transgenesis in Human Embryonic Stem Cells as a Novel Tool to Define the Human Neural Lineage," 2009, Stem Cells 27:521-532.
Placzek and Briscoe, "The Floor Plate: Multiple Cells, Multiple Signals," Nat. Rev. Neurosci., 6:230-240 (2005).
Placzek et al., "Induction of floor plate differentiation by contact-dependent, homeogenetic signals," Development, 117:205-218 (1993).
Placzek, "The role of the notochord and floor plate in inductive interactions," Curr Opin Genet Dev., 5:499-506 (1995).
Raymon et al., "Immortalized Human Dorsal Root Ganglion Cells Differentiate into Neurons with Nociceptive Properties," 1999, J. Neurosci. 19(13):5420-5428.
Reubinoff et al., "Neural progenitors from human embryonic stem cells," Nature Biotechnology, 19:1134-1140 (2001).
Roelink et al., "Floor plate and motor neuron induction by vhh-1, a vertebrate homolog of hedgehog expressed by the notochord," Cell, 76:761-775 (1994).
Saha et al., "Technical Challenges in Using Human Induced Pluripotent Stem Cells to Model Disease," Cell Stem Cell, 5(6):584-595 (2009).
Sasai et al., "Xenopus chordin: a Novel Dorsalizing Factor Activated by Organizer-Specific Homeobox Genes," Cell, 79(5):779-790 (1994).
Schlosser et al., "Development of Neurogenic Placodes in Xenopus laevis," The Journal of Comparative Neurology, 418:121-146 (2000).
Schlosser et al., "Induction and Specification of Cranial Placodes," Dev Biol., 294(2):303-351 (2006) A: pp. 303-327.
Schlosser et al., "Induction and Specification of Cranial Placodes," Dev Biol., 294(2):303-351 (2006) B: pp. 328-351.
Shen et al., "The timing of cortical neurogenesis is encoded within lineages of individual progenitor cells," Nat Neurosci., 9:743-751 (2006).
Shirasaki et al., "Guidance of cerebellofugal axons in the rat embryo: directed growth toward the floor plate and subsequent elongation along the longitudinal axis," Neuron, 14:961-972 (1995).
Smith et al., "Expression Cloning of noggin, a New Dorsalizing Factor Localized to the Spemann organizer in Xenopus Embryos," Cell, 70:829-840 (1992).
Smith et al., "Inhibition of Activin/Nodal signaling promotes specification of human embryonic stem cells into neuroectoderm," Dev Biol., 313:107-117 (2008).
Steinbeck et al., "Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson's disease model," 2015, Nature Biotechnology 33(2):204-209.
Stem Cell: Scientific Progress and Future Research Directions, Department of Health and Human Services, Chapter 4, pp. 23-42, Jun. 2001.
Streit, "Early development of the cranial sensory nervous system: from a common field to individual placodes," Dev Biol., 276:1-15 (2004).
Sukoyan et al., "Embryonic Stem Cells Derived from Morulae, Inner Cell Mass, and Blastocysts of Mink: Comparisons of Their Pluripotencies," Mol. Reprod. Dev., 36:148-158 (1993).
Sukoyan et al., "Isolation and Cultivation of Blastocyst-Derived Stem Cell Lines from American Mink (Mustela vison)," Mol. Reprod. Dev., 33:418-431 (1992).
Sumi et al., "Defining early lineage specification of human embryonic stem cells by the orchestrated balance of canonical Wnt/β-catenin, Activin/Nodal and BMP signaling," Development, 135:2969-2979 (2008).
Sun et al., "A Central Role for Islet1 in Sensory Neuron Development Linking Sensory and Spinal Gene Regulatory Programs," Nat Neurosci., 11(11):1283-1293 (2008).
Sun et al., "Design, Synthesis, and Evaluations of Substituted 3-[(3- or 4-Carboxyethylpyrrol-2-yl)Methylidenyl]indolin-2-ones As Inhibitors of VEGF, FGF, and PDGF Receptor Tyrosine Kinases," J Med Chem., 42(25):5120-5130 (1999).
Suter et al., "A Soxl to Pax6 switch drives neuroectoderm to radial glia progression during differentiation of mouse embryonic stem cells," Stem Cells, 27(1):49-58 (2009).
Takahashi et al., "Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors," Cell, 131:861-872 (2007).
Takahashi et al., "Induction of Pluripotent Stem Cells From Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors," Cell, 126(4):663-676 (2006).
Tanaka et al., "FGF-Induced Vesicular Release of Sonic Hedgehog and Retinoic Acid in Leftward Nodal Flow Is Critical for Left-Right Determination," Nature, 435(7039):172-177 (2005).
Tesar et al., "New cell lines from mouse epiblast share defining features with human embryonic stem cells," Nature, 448:196-199 (2007).
Theos et al., "The Silver Locus Product Pmell 7/Gp100/Silv/Me20: Controversial in Name and in Function," Pigment Cell Res., 18(5):322-336 (2005).
Thomson et al. (PNAS, 92:7844-7848 (Aug. 1995)). *
Tomishima et al., "Production of Green Fluorescent Protein Transgenic Embryonic Stem Cells Using the GENSAT Bacterial Artificial Chromosome," 2007, Stem Cells 25:39-45.
U.S. Appl. No. 13/201,137 (U.S. Pat. No. 8,642,334), filed Nov. 10, 2011 (Feb. 4, 2014).
U.S. Appl. No. 13/201,137, Dec. 23, 2013 Issue Fee Payment.
U.S. Appl. No. 13/201,137, Jan. 24, 2013 Response to Restriction Requirement.
U.S. Appl. No. 13/201,137, Jan. 3, 2013 Restriction Requirement.
U.S. Appl. No. 13/201,137, Jul. 24, 2013 Final Office Action.
U.S. Appl. No. 13/201,137, Jun. 6, 2013 Response to Non-Final Office Action.
U.S. Appl. No. 13/201,137, Mar. 7, 2013 Non-Final Office Action.
U.S. Appl. No. 13/201,137, Sep. 17, 2013 Response after Final Action.
U.S. Appl. No. 13/201,137, Sep. 24, 2013 Notice of Allowance.
U.S. Appl. No. 13/697,274 (U.S. Pat. No. 9,453,198), filed Jan. 22, 2013 (Sep. 27, 2016).
U.S. Appl. No. 13/697,274, Apr. 7, 2016 Request for Continued Examination (RCE).
U.S. Appl. No. 13/697,274, Aug. 19, 2016 Issue Fee Payment.
U.S. Appl. No. 13/697,274, Dec. 23, 2015 Notice of Allowance.
U.S. Appl. No. 13/697,274, Dec. 26, 2014 Restriction Requirement.
U.S. Appl. No. 13/697,274, Mar. 22, 2016 Issue Fee Payment.
U.S. Appl. No. 13/697,274, Mar. 24, 2016 Applicant Initiated Interview Summary.
U.S. Appl. No. 13/697,274, Mar. 24, 2016 Notice of Allowance.
U.S. Appl. No. 13/697,274, Mar. 26, 2015 Response to Restriction Requirement.
U.S. Appl. No. 13/697,274, May 20, 2016 Notice of Allowance.
U.S. Appl. No. 13/697,274, May 5, 2015 Non-Final Office Action.
U.S. Appl. No. 13/697,274, Nov. 4, 2015 Response to Non-Final Office Action.
U.S. Appl. No. 14/168,835 (US 2017/0130199), filed Jan. 30, 2014 (May 11, 2017).
U.S. Appl. No. 14/168,835, Apr. 10, 2018 Advisory Action.
U.S. Appl. No. 14/168,835, Apr. 16, 2018 Applicant Initiated Interview Summary.
U.S. Appl. No. 14/168,835, Apr. 16, 2018 Request for Continued Examination (RCE).
U.S. Appl. No. 14/168,835, Apr. 25, 2018 Non-Final Office Action.
U.S. Appl. No. 14/168,835, Jan. 17, 2018 Final Office Action.
U.S. Appl. No. 14/168,835, Jan. 27, 2017 Office of Petitions Decision.
U.S. Appl. No. 14/168,835, Mar. 15, 2016 Office of Petitions Decision.
U.S. Appl. No. 14/168,835, Mar. 16, 2018 Response after Final Action.
U.S. Appl. No. 14/168,835, May 22, 2017 Non-Final Office Action.
U.S. Appl. No. 14/168,835, Nov. 16, 2017 Response to Non-Final Office Action.
U.S. Appl. No. 14/168,835, Nov. 26, 2014 Petition for review by the Office of Petitions.
U.S. Appl. No. 14/168,835, Sep. 15, 2016 Petition for review by the Office of Petitions.
U.S. Appl. No. 14/168,835, Sep. 30, 2014 Notice of Abandonment.
U.S. Appl. No. 14/169,286 (US 2017/0159012), filed Jan. 31, 2014 (Jun. 8, 2017).
U.S. Appl. No. 14/169,286, Aug. 8, 2017 Response to Restriction Requirement.
U.S. Appl. No. 14/169,286, Dec. 4, 2017 Response to Non-Final Office Action.
U.S. Appl. No. 14/169,286, Feb. 12, 2018 Non-Final Office Action.
U.S. Appl. No. 14/169,286, Jun. 8, 2017 Restriction Requirement.
U.S. Appl. No. 14/169,286, Mar. 15, 2016 Office of Petitions Decision.
U.S. Appl. No. 14/169,286, Mar. 3, 2017 Office of Petitions Decision.
U.S. Appl. No. 14/169,286, May 11, 2018 Response to Non-Final Office Action.
U.S. Appl. No. 14/169,286, Nov. 26, 2014 Petition for review by the Office of Petitions.
U.S. Appl. No. 14/169,286, Sep. 15, 2016 Petition for review by the Office of Petitions.
U.S. Appl. No. 14/169,286, Sep. 30, 2014 Notice of Abandonment.
U.S. Appl. No. 14/169,286, Sep. 6, 2017 Non-Final Office Action.
U.S. Appl. No. 14/922,110 (US 2016/0115448), filed Oct. 23, 2015 (Apr. 28, 2016).
U.S. Appl. No. 14/922,110, Jan. 3, 2018 Response to Restriction Requirement.
U.S. Appl. No. 14/922,110, Jul. 10, 2017 Restriction Requirement.
U.S. Appl. No. 14/922,110, May 17, 2018 Non-Final Office Action.
U.S. Appl. No. 15/077,012 (US 2016/0201032), filed Mar. 22, 2016 (Jul. 14, 2016).
U.S. Appl. No. 15/077,012, Jan. 16, 2018 Response to Restriction Requirement.
U.S. Appl. No. 15/077,012, Jun. 12, 2017 Non-Final Office Action.
U.S. Appl. No. 15/077,012, Mar. 22, 2018 Notice of Allowance.
U.S. Appl. No. 15/077,012, Nov. 3, 2017 Restriction Requirement.
U.S. Appl. No. 15/077,012, Sep. 12, 2017 Response to Non-Final Office Action.
Valenzuela et al., "Identification of Mammalian Noggin and Its Expression in the Adult Nervous System," J. Neurosci., 15(9):6077-6084 (1995).
Vallier et al., "Nodal Inhibits Differentiation of Human Embryonic Stem Cells Along the Neuroectodermal Default Pathway," Dev Biol., 275(2):403-421 (2004).
Venezia et al., "Molecular Signatures of Proliferation and Quiescence in Hematopoietic Stem Cells," PLoS Biol., 2(10):e301 (2004).
Vierbuchen et al., "Direct Conversion of Fibroblasts to Functional Neurons by Defined Factors," Nature, 463(7284):1035-1041 (2010).
Wang (2010, PNAS, 107:9323-9328). *
Wang et al., "Noggin and bFGF cooperate to maintain the pluripotency of human embryonic stem cells in the absence of feeder layers," Biochem Biophys Res Commun., 330:934-942 (2005).
Wang, Stem Cells and Development, 2010, 19:1375-1383. *
Watanabe et al., "A ROCK inhibitor permits survival of dissociated human embryonic stem cells," Nat. Biotechnol., 25(6):681-686 (2007).
Watanabe et al., "Directed differentiation of telencephalic precursors from embryonic stem cells," Nat. Neuro., 3:288-296 (2005).
Weinstein et al., "Neural Induction," Annu Rev Cell Dev Biol., 15:411-433, pp. 411-424 (1999a).
Weinstein et al., "Neural Induction," Annu Rev Cell Dev Biol., 15:411-433, pp. 425-433 (1999b).
Wichterle et al., "Directed Differentiation of Embryonic Stem Cells into Motor Neurons," Cell, 110:385-397 (2002).
Willert, CSH Perspectives in Biology, 2012, 4:1-13. *
Woolf et al., "Nociceptors-Noxious Stimulus Detectors" 2007, Neuron 55:353-364.
Written Opinion of the International Searching Authority dated Mar. 29, 2013 in International Patent Application No. PCT/US2012/063339.
Xu et al., "Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells," Nat Methods, 2:185-190 (2005).
Xu et al., "BMP4 initiates human embryonic stem cell differentiation to trophoblast," Nat Biotechnol., 20(12):1261 (2002).
Xu et al., "NANOG is a Direct Target of TGFβ/Activin-Mediated SMAD Signaling in Human ESCs," Cell Stem Cell, 3:196-206 (2008).
Yamashita et al., "Bone Morphogenetic Protein Receptors," Bone, 19(6):569-574 (1996).
Yan et al., "Directed Differentiation of Dopaminergic Neuronal Subtypes from Human Embryonic Stem Cells," Stem Cells, 23(6):781-790 (2005).
Yu et al., "BMP type I receptor inhibition reduces heterotopic ossification", Nature Medicine 14(12):1363-1369 (2008).
Zhang and Zhang, "Differentiation of Neural Precursors and Dopaminergic Neurons from Human Embryonic Stem Cells," Methods Mal Biol., 584:355-366 (2010).
Zhang et al., "In vitro differentiation of transplantable neural precursors from human embryonic stem cells," Nature Biotechnology, 19:1129-1133 (2001).
Zhou et al., "High-Efficiency Induction of Neural Conversion in Human ESCs and Human Induced Pluripotent Stem Cells With a Single Chemical Inhibitor of Transforming Growth Factor Beta Superfamily Receptors," Stem Cells, 28(10):1741-1750 (2010).
Zhu et al., "Functional Smoothened Is Required for Expression of Gli3 in Colorectal Carcinoma Cells," Cancer Letters, 207(2):205-214 (2004).
Zietlow et al., "The Survival of Neural Precursor Cell Grafts Is Influenced by In Vitro Expansion," Journal of Anatomy, 207(3):227-240 (2005).
Zoltewicz et al., "oto is a homeotic locus with a role in anteroposterior development that is partially redundant with Lim1," Development, 126:5085-5095 (1999).

Also Published As

Publication number Publication date
BR112014010776A8 (pt) 2020-03-17
BR112014010776B1 (pt) 2021-11-30
IL273910A (en) 2020-05-31
AU2021218116A1 (en) 2021-09-09
ES2779453T3 (es) 2020-08-17
AU2018222997B2 (en) 2021-05-20
IL295556A (en) 2022-10-01
WO2013067362A1 (fr) 2013-05-10
AU2012332269B2 (en) 2018-05-31
IL232450A0 (en) 2014-06-30
EP3693455A1 (fr) 2020-08-12
JP2020178695A (ja) 2020-11-05
JP6461602B2 (ja) 2019-01-30
KR20200061414A (ko) 2020-06-02
PL2773748T3 (pl) 2020-06-15
DK2773748T3 (da) 2020-03-16
EP2773748A1 (fr) 2014-09-10
CN110684734A (zh) 2020-01-14
BR122020000182B8 (pt) 2022-05-24
JP2017212989A (ja) 2017-12-07
AU2018222997A1 (en) 2018-09-20
KR20220108208A (ko) 2022-08-02
EA201490796A1 (ru) 2014-10-30
JP7225163B2 (ja) 2023-02-20
CN110684733A (zh) 2020-01-14
JP6756786B2 (ja) 2020-09-16
BR112014010776A2 (pt) 2017-04-25
IL286793A (en) 2021-10-31
JP6588500B2 (ja) 2019-10-09
AU2012332269A1 (en) 2014-05-22
US20190211306A1 (en) 2019-07-11
US11970712B2 (en) 2024-04-30
CA3177246A1 (fr) 2013-05-10
IL232450B (en) 2020-04-30
EP2773748B1 (fr) 2019-12-25
CN113481159A (zh) 2021-10-08
KR102426327B1 (ko) 2022-08-01
BR122020000182B1 (pt) 2022-04-19
PT2773748T (pt) 2020-03-26
JP2019010104A (ja) 2019-01-24
US20150010514A1 (en) 2015-01-08
JP2023027064A (ja) 2023-03-01
CA2854578A1 (fr) 2013-05-10
KR20140116063A (ko) 2014-10-01
CN104379731A (zh) 2015-02-25
CN113528442A (zh) 2021-10-22
IL286793B (en) 2022-09-01
HK1201879A1 (en) 2015-09-11
JP2014532440A (ja) 2014-12-08
JP2021000104A (ja) 2021-01-07
US20200407680A1 (en) 2020-12-31
KR20230136702A (ko) 2023-09-26
CA2854578C (fr) 2023-01-03
KR102581669B1 (ko) 2023-09-22
KR102115273B1 (ko) 2020-05-28
EP2773748A4 (fr) 2015-04-08
US10711243B2 (en) 2020-07-14
IL273910B (en) 2021-10-31

Similar Documents

Publication Publication Date Title
US11970712B2 (en) Midbrain dopamine (DA) neurons for engraftment
JP2022088676A (ja) 幹細胞由来外胚葉系統前駆体を分化する方法
JP2023075336A (ja) 幹細胞由来外胚葉系統前駆体を分化する方法
EA041083B1 (ru) Способ индукции дифференцировки плюрипотентных стволовых клеток in vitro

Legal Events

Date Code Title Description
AS Assignment

Owner name: MEMORIAL SLOAN-KETTERING CANCER CENTER, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STUDER, LORENZ;SHIM, JAE-WON;KRIKS, SONJA;SIGNING DATES FROM 20140716 TO 20140721;REEL/FRAME:033441/0788

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:SLOAN-KETTERING INST CAN RESEARCH;REEL/FRAME:034495/0197

Effective date: 20141014

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4